WO2012016203A1 - Chimeric il-1 receptor type i agonists and antagonists - Google Patents

Chimeric il-1 receptor type i agonists and antagonists Download PDF

Info

Publication number
WO2012016203A1
WO2012016203A1 PCT/US2011/045995 US2011045995W WO2012016203A1 WO 2012016203 A1 WO2012016203 A1 WO 2012016203A1 US 2011045995 W US2011045995 W US 2011045995W WO 2012016203 A1 WO2012016203 A1 WO 2012016203A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
residues
cytokine
domain
segments
Prior art date
Application number
PCT/US2011/045995
Other languages
French (fr)
Inventor
Thomas M. Barnes
Jinzhao Hou
Bracken M. King
Original Assignee
Eleven Biotherapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to JP2013522011A priority Critical patent/JP6030554B2/en
Priority to MX2013001107A priority patent/MX360097B/en
Application filed by Eleven Biotherapeutics, Inc. filed Critical Eleven Biotherapeutics, Inc.
Priority to CN201180047508.1A priority patent/CN103221422B/en
Priority to EP23157789.1A priority patent/EP4219538A3/en
Priority to SG2013006697A priority patent/SG187231A1/en
Priority to EP11749612.5A priority patent/EP2598526B1/en
Priority to US13/812,582 priority patent/US8853150B2/en
Priority to DK11749612.5T priority patent/DK2598526T3/en
Priority to RU2013108836/10A priority patent/RU2013108836A/en
Priority to KR1020137005337A priority patent/KR102080704B1/en
Priority to PL11749612T priority patent/PL2598526T3/en
Priority to BR112013003646-0A priority patent/BR112013003646B1/en
Priority to NZ605928A priority patent/NZ605928A/en
Priority to SI201131571T priority patent/SI2598526T1/en
Priority to ES11749612T priority patent/ES2701405T3/en
Priority to LTEP11749612.5T priority patent/LT2598526T/en
Priority to CA2806438A priority patent/CA2806438C/en
Priority to AU2011283669A priority patent/AU2011283669A1/en
Priority to EP18191564.6A priority patent/EP3470421B9/en
Publication of WO2012016203A1 publication Critical patent/WO2012016203A1/en
Priority to IL224435A priority patent/IL224435B/en
Priority to US14/460,563 priority patent/US9458216B2/en
Priority to AU2016213845A priority patent/AU2016213845B2/en
Priority to IL259547A priority patent/IL259547B/en
Priority to HRP20181446TT priority patent/HRP20181446T1/en
Priority to AU2018250431A priority patent/AU2018250431A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/545IL-1
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/14Decongestants or antiallergics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • IL-1 a and IL-1 ⁇ are prototypic members of a family of immunoregulatory cytokines and have several prominent roles in regulating the immune system.
  • IL-1 a and IL-1 ⁇ bind to the interleukin-1 receptor I (IL-1 Rl), leading to the engagement of the secondary receptor, interleukin-1 receptor accessory protein (IL- 1 RAcP).
  • IL-1 Rl interleukin-1 receptor I
  • IL- 1 RAcP interleukin-1 receptor accessory protein
  • Signaling agonized by IL-1 a and IL-1 ⁇ leads to amplified T cell responses, including the proliferation and survival of na ' ive T cells and the development of T H 17 cells.
  • cytokine domains that can be used, inter alia, to modulate cellular signalling responsive to interleukin-1 receptor I (IL-1 Rl), to treat disorders, and to detect and/or bind to cellular receptors, as well as other agents.
  • IL-1 Rl interleukin-1 receptor I
  • this disclosure features an isolated protein including an cytokine domain that contains amino acid residues from at least two parental cytokines domains, for example, receptor binding features, surface features, ⁇ strands, and loops from at least two parental cytokines domains.
  • the cytokine domain binds to IL-1 Rl and includes receptor binding features from different parental cytokine domains, e.g., from a receptor agonist and a receptor antagonist (such as IL-1 ⁇ and IL-1 Ra, or IL-1 a and IL-1 Ra), from IL-1 ⁇ and IL-1 a, or from all three of IL-1 Ra, IL-1 a and IL-1 Ra.
  • the receptor binding features can correspond to residues, segments, or regions in Sites A and B. With respect to such residues, segments, and regions corresponding to Sites A and B, in the context of IL-1 (IL-1 ⁇ , IL-1 a, and IL-1 Ra), see the definitions further below.
  • the cytokine domain may have: (a)(i) Site A residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in a first parental cytokine domain; (a)(ii) Extended Site A residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in a first parental cytokine domain; (a)(iii) Site A segments A1 and A2 that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding regions of a first parental cytokine domain; and/or (a)(iv) a Site A region that is at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding regions of a first parental cytokine domain.
  • the cytokine domain may have: (b)(i) Site B residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in a second parental cytokine domain; (b)(ii) extended Site Bs residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in a second parental cytokine domain; (b)(iii) Site B segments B1 , B2, and B3 that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding regions of a second parental cytokine domain; and/or (b)(iv) a Site B region that is at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding regions of a second parental cytokine domain.
  • the cytokine domain includes features: (a)(i) and b(i), (a)(ii) and b(ii), (a)(iii) and (b)(iii), or (a)(iv) and (b)(iv), e.g., wherein each feature is further defined by 80, 85, 88, 90, 92, 95, or 100% identity.
  • the first parental cytokine domain can be I L- 1 ⁇
  • the second parental cytokine domain can be IL-1 Ra.
  • the first parental cytokine domain can be IL-1 a
  • the second parental cytokine domain can be IL-1 Ra.
  • the cytokine domain can also include amino acids from a second parental cytokine domain at one or more positions in the domain that impair interaction with a cytokine secondary receptor (e.g., IL-1 RAcP).
  • a cytokine secondary receptor e.g., IL-1 RAcP
  • the second parental cytokine domain is IL-1 Ra.
  • the cytokine domain includes one or more Site C and/or D segments (e.g., C1 , D1 , D2, D3, D4, and/or D5) from IL-1 Ra, or sequences at least 80, 85, 88, 90, 92, 95, or 100% identical to such segments.
  • the cytokine domain includes (i) a Site C residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra, (ii) Site D residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra, (iii) a C1 segment that is at least 70, 75, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra; or (iv) a D2 segment that is identical at at least 3, 4, or 5 residues to corresponding residues in IL- 1 Ra.
  • the cytokine domain can include features (i) and (ii), or (ii) and (iii), e.g., wherein each feature is further defined by 80, 85, 88, 90, 92, 95, or 100% identity, or (iii) and (iv)
  • the domain can include regions from at least two different human IL-1 family cytokine domains, wherein the regions are selected from the group consisting of the A region (having A1 and A2 segments), the B region (having B1 , B2, and B3 segments), the C region, and the D region (having D1 , D2, D3, D4, and D5 segments).
  • the cytokine domain can include a Site A region and a Site B region from different cytokine domains.
  • the Site A region can be from a naturally occuring receptor agonist or antagonist; the Site B region can be from a naturally occuring receptor agonist. It can include a Site C region from a naturally occuring receptor antagonist and/or a Site D region from a naturally occuring receptor antagonist.
  • the domain can be a chimeric domain having segments that are at least 5, 6, 10, 15, 20, or 25 amino acids in length and that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding segments from at least two different parental cytokine domains, such as a first and second parental cytokine domain.
  • the parental cytokine domains can be IL-1 Rl binding cytokines, such as IL-1 ⁇ , IL-1 a, and IL-1 Ra.
  • the amino acids that are not in the segments from the first parental cytokine domain are from two or more other parental cytokine domains.
  • the cytokine domain includes at least two segments of at least 5, 6, 10, 15, 20, or 25 amino acids in length that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding segments of a first parental cytokine domains, and the amino acids that are not in such segments are predominantly (e.g., at least 50, 60, 70, 80, 85, 88, 90, 92, 95, or 100%) identical to corresponding residues in the second parental cytokine domain.
  • the cytokine domain includes (i) at least two segments of at least 5, 6, 10, 15, 20, or 25 amino acids in length that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding segments of a first parental cytokine domains, and (ii) at least one, two or three segments, e.g., of at least 5, 6, 7, 8, 10, or 15 amino acids in length, that are identical to a second parental cytokine domain.
  • the cytokine domain can include a first segment of 20-50, 25-50, 30-45, or 30-40 amino acids (e.g., 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, or 40), and a second segment of 20-45, 20-40, 25-40, or 25-35 amino acids (e.g., 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, or 35), each identical (or at least 80, 85, 88, 90, 92, 95, or 98% identical) to a first parental cytokine domain (e.g., IL-1 Ra), and a third segment that is identical (or at least 80, 85, 88, 90, 92, 95, or 98% identical) to a second parental cytokine domain (e.g., IL-1 ⁇ or IL-1 a).
  • first parental cytokine domain e.g., IL-1 Ra
  • a third segment that is identical (or at least 80, 85, 88, 90, 92, 95,
  • the third segment can be between 55-90, 60-90, 60-85, or 70-85 amino acids in length, e.g., 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, or 85 amino acids in length.
  • the first segment can be a segment at least 80, 85, 88, 90, 92, 95, 98, or 100% to WDVNQKTFYLRNNQLVAGYLQGPNV (SEQ ID NO:9, also termed the A1 segment herein, and corresponding to residues 16-40 of SEQ ID NO:3 and residues 1 1 -36 according to IL-1 ⁇ numbering).
  • the second segment can be a segment at least 80, 85, 88, 90, 92, 95, 98, or 100% to residues 120-140 or 120-141 of SEQ ID NO:3 (IL-1 Ra), corresponding to residues 121 -139 or 121 -140 (according to IL- 1 ⁇ numbering).
  • the third segment can be at least 80, 85, 88, 90, 92, 95, 98, or 100% to residues 45-100 or 42-120 from IL-1 ⁇ (SEQ ID NO:1 ).
  • the first segment can be a segment at least 80, 85, 88, 90, 92, 95, 98, or 100% to residues 14-45 of SEQ ID NO:3 (corresponding to residues 9-41 according to IL-1 ⁇ numbering).
  • the second segment can be a segment at least 80, 85, 88, 90, 92, 95, 98, or 100% to residues 120-145 of SEQ ID NO:3 (corresponding to residues 121 -145 according to IL- ⁇ ⁇ numbering) or residues 120-147 of SEQ ID NO:3 (corresponding to residues 121 -147 according to I L- 1 ⁇ numbering).
  • At least residues 1 1 -41 and 120-147 are collectively at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra.
  • a terminus of one of the segments of the first IL-1 family cytokine domain is located within five, four, three, two, or one amino acids of amino acid 41 of SEQ ID NO:1 and a terminus of one of the segments of the first IL-1 family cytokine domain is located within five, four, three, two, or one amino acids of amino acid 121 of SEQ ID NO:1 .
  • the domain includes a segment having an N-terminus at a position within five, four, three, two, or one amino acids of amino acid 42 of SEQ ID NO:1 and a C-terminus within five, four, three, two, or one amino acids of amino acid 120 of SEQ ID NO:1 , and/or a segment having an N-terminus at a position within five, four, three, two, or one amino acids of amino acid 121 of SEQ ID NO:1 and a C-terminus within five, four, three, two, or one amino acids of amino acid 145 of SEQ ID NO:1 .
  • residues in the domain at positions corresponding to 1 1 -41 and 120-147 are collectively at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra.
  • the domain can also be based on sequences from IL-1 cytokine family members, e.g., at analogous positions to the foregoing.
  • the cytokine domain includes one, two, three or more of the following sequences: WDVNQKTFYLRNNQLVAGYLQGPNV (SEQ ID NO:9);
  • NLEEK (SEQ ID NO:10); RIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEK(SEQ ID NO:1 1 ); AMEADQP(SEQ ID NO:12); FLCTAMEADQPVSLTNMPDEGVMVTKFY(SEQ ID NO:13); and/or sequences at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to the foregoing.
  • the cytokine domain includes one, two, three or more of the following sequences: VQGEESNDKI (SEQ ID NO:14); KKKMEKRF (SEQ ID NO:15); and FSMSFVQGEESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKN YPKKKMEKRFVFNKIEINNKLEFES (SEQ ID NO:16); and/or sequences at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to the foregoing.
  • one or more or all of the ⁇ 1 ⁇ 2, ⁇ 2 ⁇ 3, ⁇ 8 ⁇ 9 and ⁇ 10 ⁇ 1 1 loops are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding loops from an IL-1 antagonist, e.g., IL-1 Ra.
  • an IL-1 antagonist e.g., IL-1 Ra.
  • one or more or all of the ⁇ 4 ⁇ 5, ⁇ 5 ⁇ 6, ⁇ 6 ⁇ 7 and ⁇ 7 ⁇ 8 loops are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding loops from a different IL-1 family cytokine domain than the human parental cytokine that is most similar to the ⁇ 1 ⁇ 2, ⁇ 2 ⁇ 3, ⁇ 8 ⁇ 9 and ⁇ 10 ⁇ 1 1 loops.
  • one or more or all of the ⁇ 4 ⁇ 5, ⁇ 5 ⁇ 6, ⁇ 6 ⁇ 7 and ⁇ 7 ⁇ 8 loops are at least 80, 85, 88, 90, 92, 95, or 100% identical to such loops from an IL-1 agonist such as I L- 1 ⁇ .
  • the ⁇ 1 1 ⁇ 12 loop is at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding loop from an IL-1 antagonist, e.g., IL-1 Ra.
  • the cytokine domain includes sequences that are at least 80, 85, 88, 90, 92, 95, or 100% identical to one, two, three, or all of beta strands ⁇ 2, ⁇ 3, ⁇ 10 and ⁇ 1 1 of IL-1 Ra. In some embodiments, the cytokine domain includes sequences that are at least 80, 85, 88, 90, 92, 95, or 100% identical to one, two, three, or all of ⁇ 4, ⁇ 6, ⁇ 7, and ⁇ 8 of IL-1 ⁇ , or to ⁇ 4, ⁇ 5, ⁇ 6, ⁇ 7, and ⁇ 8 of IL-1 ⁇ .
  • the cytokine domain does not contain a segment that is greater than 80, 85, 90, 95, or 100% identical to amino acids I46-G59, A55-G59, A55- V83, I60-V83, N84-D95, I46-S1 10, V49-S1 10, or I46-G1 18 of SEQ ID NO:3. In some embodiments, the cytokine domain does not contain a segment that is greater than 80, 85, 90, 95, or 100% identical to amino acids N7-V41 , R1 1 -M36, N102-D145, or Y121 - D145 of SEQ ID NO:1 .
  • the cytokine domain is not naturally occurring. It differs from human IL-1 family cytokine domains. For example, it is less than 98, 95, 90, 85, 80, 75, 70, 65, 60, or 55% identical to IL-1 Ra (SEQ ID NO:3), IL-1 ⁇ (SEQ ID NO:1 ), and/or IL-1 a (SEQ ID NO:2).
  • the cytokine domain can also be at least 30, 40, 45, 50, 55, 60, 65, 70% identical to such cytokine.
  • the chimeric domain can be between 30-95%, 40-90%, or 45-85% identical to IL-1 Ra, IL-1 ⁇ , and IL-1 a.
  • the chimeric domain can be between 40-90% identical to IL-1 ⁇ and 35-85% identical to IL-1 Ra;
  • the chimeric domain can be between 40-90% identical to IL-1 a and 35-85% identical to IL-1 Ra; between 40-80% identical to IL-1 a and 45-80% identical to IL-1 Ra; between 45-72% identical to IL-1 a and 45-80% identical to IL-1 Ra; between 45-72% identical to IL-1 a and 53-80% identical to IL-1 Ra; between 50-72% identical to IL-1 a and 53-70% identical to IL-1 Ra; between 60- 72% identical to IL-1 a and 53-68% identical to IL-1 Ra; between 65-72% identical to IL-1 a and 54-60% identical to IL-1 Ra; or between 68-72% identical to IL-1 a and 54-57% identical to IL-1 Ra.
  • the cytokine domain differs from IL-1 Ra, and binds to the receptor while including a Site C and/or Site D characteristic of a naturally occurring receptor antagonist (such as IL-1 Ra).
  • the domain is less than 98, 95, 92, 90, 85, and 80% identical from human IL-1 ⁇ and/ IL-1 Ra.
  • the domain is between 40-95%, 40-90% or 45-85% identical to IL-1 Ra.
  • the domain can also be between 40-95%, 40-90% or 45-85% to an IL-1 family cytokine agonist, such as IL-1 a or I L- 1 ⁇ .
  • the cytokine domain includes: at least 40, 45, 50, 55, 60, 65, 70, 75, 80 or 85% amino acids from a first parental cytokine domain that is an agonist of cytokine signaling.
  • the cytokine domain has greater amino acid identity (e.g., at least 5, 10, 15, or 20% greater) to a receptor agonist (such as IL-1 ⁇ or IL-1 a) than to a receptor antagonist (IL-1 Ra), but functions as an antagonist of IL-1 Rl.
  • the cytokine domain is completely chimeric, e.g., each amino acid in the domain is from one of the parental cytokine domains, e.g., one of two parental cytokine domains or one of three or more parental cytokine domains.
  • the parental cytokine domains are human cytokine domains or non-human primate cytokine domains.
  • the cytokine domain is partially chimeric, e.g., not all amino acids in the domain are from one of the parental cytokine domains.
  • the isolated protein binds to IL-1 Rl and modulates signaling by the receptor, e.g., agonizes or antagonizes IL-1 Rl receptor signaling activity.
  • the protein does not substantially induce IL-6 production when contacted to IL-1 ⁇ responsive human cells and/or does not substantially induce production of an IL-1 ⁇ responsive reporter gene, e.g., at concentrations of 10 g ml, 100 g ml, or 1 mg/ml.
  • the protein inhibits signaling by IL-1 ⁇ (e.g., at a concentration of 0.1 ng/ml such as in a cellular assay described herein) with an IC50 of less than 100, 50, 20, 10, or 5 nM.
  • the protein can inhibit signaling by IL-1 ⁇ with an IC50 that is less than that of IL-1 Ra, e.g., at least 10, 20, or 50% lower.
  • the cytokine domain binds to e.g., with the same or better affinity than one of the parental cytokine domains. In some embodiments, the cytokine domain binds to IL-1 Rl with K D of less than 100, 50, 20, 10, 5, or 1 nM, or less than 500, 400, 100, or 50 pM.
  • the association constant can be greater than 1 x 1 0 4 , 3 ⁇ 1 0 4 , 1 ⁇ 1 0 5 , or 1 ⁇ 10 6 M " V, and the dissociation can be less than 1 x 10 "3 , 1 x 1 0 "4 , 6x 1 0 "4 , or 6x 1 0 "5 S "1 .
  • the cytokine domain binds to IL-1 Rl with a better affinity (e.g., lower K D ) and/or a slower dissociation rate than IL-1 ⁇ or IL-1 Ra.
  • the cytokine domain can bind to IL-1 Rl with a dissociation constant less than or equal to that of IL-1 Ra and/or with an association constant greater or equal to that of IL-1 ⁇ .
  • the cytokine domain can be between approximately 120-180, 140-170, or 148-
  • the domain is 152 or 153 amino acids in length.
  • the domain includes at least 10, 1 1 , or 12 ⁇ -strands, and is stably folded.
  • the cytokine domain has a Tm of at least 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, or 64°C, as described herein. It can have a Tm of between 51 -61 , 51 -66, 56-61 , or 56-66°C.
  • the cytokine domain does not begin unfolding until at least 48, 50, 51 , 55, 57, 58, or 59°C.
  • the domain can have a Tm that is at least 2, 4, 6, 7, or 8°C greater than the Tm of IL-1 Ra and/or IL-1 ⁇ in a physiological buffer, e.g., between about 5-12, 5-10, or 7-10°C greater than the Tm of IL-1 Ra and/or IL-1 ⁇ at a concentration of about 0.5 mg/ml.
  • the protein can include other features described herein.
  • the disclosure features an isolated protein that includes a chimeric IL-1 family cytokine domain.
  • IL-1 cytokine family members include IL-1 a, IL-1 ⁇ , IL-1 Ra, IL-18, IL-1 F5, IL-1 F6, IL-1 F7, IL-1 F8, IL-1 F9, IL-1 F10, and IL-33.
  • the cytokine domain can include a receptor binding region of one of the foregoing cytokines or a protein sequence that includes elements of one or more such cytokines.
  • the cytokine domain can include a chimera of two or more IL-1 cytokine family members.
  • the chimeric domain includes at least one segment of length at least five, six, seven, eight, nine, ten, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, or 70 amino acids and having amino acid identity to (or at least 80, 82, 85, 87, 90, 92, 94, 95, 96, 97, 98, or 99% identity to) to a first IL-1 family cytokine, and another segment of length of length at least five, six, seven, eight, nine, ten, 15, 20, 25, 30, 35, or 40 amino acids and having amino acid identity to (or at least 80, 82, 85, 87, 90, 92, 94, 95, 96, 97, 98, or 99% identity to) to a second IL-1 family cytokine.
  • the chimeric domain can be less than 90, 85, 80, or 75% identical to one or both of the first and second IL-1 family cytokine.
  • the first and second IL-1 family cytokines are selected from the group consisting of IL-1 ⁇ , IL-1 a, and IL-1 Ra. In another embodiment, the first and second IL-1 family cytokines are selected from the group consisting of IL-1 F5, IL-1 F6, IL-1 F7, and IL-1 F8. In another embodiment, the first IL-1 family cytokine is selected from the group of agonists and the second is selected from the group of antagonists. In some embodiments, the chimeric domain includes fewer than 120, 1 10, 100, 90, or 80 contiguous amino acids from the same parental cytokine domain.
  • the chimeric domain is identical to the first IL-1 family cytokine at at least 50, 60, 70, 80, 90, 100, 1 10, or 120 positions and is identical to the second IL-1 family cytokine at at least 50, 60, 70, 80, 90, 100, 1 10, or 120 positions (including positions that may individually be identical to both such cytokines).
  • the chimeric domain includes at least two, three, or four discontinuous segments, each of length at least five, six, seven, eight, nine, ten, 15, or 20 amino acids and having amino acid identity to (or at least 80, 82, 85, 87, 90, 92, 94, 95, 96, 97, 98, or 99% identity to) corresponding segments of a first IL-1 family cytokine and including amino acids predominantly from a second IL-1 family cytokine at remaining positions.
  • the chimeric domain includes 4, 5, 6, or 7 segments, wherein adjacent segments are from different parental IL-1 family cytokine domains.
  • each amino acid in the domain is located in a peptide of at least 5 or 6 amino acids in length from a naturally occurring human IL-1 family cytokine domain.
  • the chimeric domain includes at least one, two, or three of: (i) a segment of at least 50, 60, 65, 70, or 75 amino acids in length from IL-1 ⁇ , (ii) a segment of at least 15, 20, 25 amino acids in length from IL-1 Ra; and (iii) another segment of at least 15, 20, 25 amino acids in length from IL-1 Ra.
  • the discontinuous segments includes residues (i) 1 -6 and 45-61 , (ii) 1 -6 and 86-95, (iii) 45-61 and 86-95, (iv) 1 -6 and 148-153, (v) 45-61 and 148- 153, or (vi) 86-95 and 148-153, according to the numbering of such positions in
  • the three discontinuous segments from the first IL-1 family cytokine can include, e.g., residues 1 -8, 42-120, and 141 -153, residues 1 -10, 37-125, and 131 -153, or residues 1 -6, 45-61 , 86-95, and 148-153, according to the numbering of such positions in IL-1 ⁇ .
  • the chimeric domain can be at least 80, 82, 85, 87, 90, 92, 94, 95, 96, 97, 98, 99, or 100% identical to the second IL-1 family cytokine at the remaining positions.
  • one or more of the borders of the discontinuous segments are located at positions wherein the first and second IL-1 family cytokines are identical or are conserved.
  • the protein can have other features described herein.
  • this disclosure features an isolated IL-1 inhibitor including a IL- 1 family cytokine domain that binds to IL-1 Rl.
  • the IL-1 inhibitor includes one or more features above or elsewhere herein.
  • the cytokine domain includes: (a) amino acids identical to IL-1 Ra at the following positions ARG1 1 , SER13, GLN14, GLN15, GLU25, LYS27, LEU29, HIS30, LEU31 , GLN32, GLY33, GLN34, ASP35, MET36, GLN38, GLN39, ALA127, GLU128, ASN129, MET130, and GLN141 (according to the numbering of IL-1 ⁇ ) and (b) amino acids identical to IL-1 ⁇ at the following positions: ALA1 , PR02, VAL3, ARG4, LEU6, PHE46, GLN48, GLU51 , SER52, ASN53, LYS55,
  • Site A segments A1 and A2 are at least 80% identical (collectively) to corresponding segments of IL-1 Ra.
  • Site B segments B1 , B2, and B3 are at least 80% identical (collectively) to corresponding segments of IL-1 ⁇ .
  • Site A segments A1 and A2 are at least 90% identical (collectively) to corresponding segments of IL-1 Ra; and
  • Site B segments B1 , B2, and B3 are at least 90% identical (collectively) to corresponding segments of IL-1 ⁇ .
  • Site A segments A1 and A2 are identical to
  • the cytokine domain includes sequences that are at least 80% identical (collectively) to beta strands ⁇ 2, ⁇ 3, ⁇ 10 and ⁇ 1 1 of IL-1 Ra and sequences that are at least 80% identical (collectively) to beta strands ⁇ 4, ⁇ 6, ⁇ 7, and ⁇ 8 of IL-1 ⁇ .
  • the cytokine domain includes sequences that are identical to beta strands ⁇ 2, ⁇ 3, ⁇ 10 and ⁇ 1 1 of IL-1 Ra and sequences that are identical to beta strands ⁇ 4, ⁇ 6, ⁇ 7, and ⁇ of IL-1 ⁇ .
  • the segments and features identified above from IL-1 ⁇ are derived from IL-1 a, or a combination of IL-1 ⁇ or IL-1 a.
  • the IL-1 inhibitor includes one or more (e.g., at least two, three, four, five, six, or seven) of the following properties: (i) Site A or Site B residues (and/or extended Site A and extended Site B residues) that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 ⁇ , IL-1 a, or IL- 1 Ra; (ii) A1 and A2 segments that are collectively at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra; (iii) B1 , B2, and B3 segments that are collectively at least 80% identical to corresponding residues in IL-1 ⁇ or IL-1 a; (iv) a Site A region that is at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to
  • a Site B region is at least 80, 85, 88, 90, 92, 95, 98, or 100% to corresponding residues in IL-1 ⁇ or IL-1 a;
  • a Site C and/or Site D residues that are at least 50, 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra or IL 36Ra,
  • a C1 segment that is at least 70, 75, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra;
  • a D2 segment that is identical at at least 3, 4, or 5 residues to corresponding residues in IL-1 Ra.
  • the cytokine domain differs from IL-1 Ra, for example, the cytokine domain is less than 99, 98, 95, 90, 85, 80, 75, 70, 65, 60% identical to IL-1 Ra, and/or at least 30, 40, 45, 50, 55, 60, 65, 70% identical to IL-1 Ra, e.g., between 40-95%, 40-90%, or 45- 85% identical to IL-1 Ra.
  • the cytokine domain does not contain a segment that is greater than 80, 85, 90, 95, or 100% identical to amino acids I46-G59, A55-G59, A55-V83, I60-V83, N84-D95, I46-S1 10, V49-S1 10, or I46-G1 18 of SEQ ID NO:3. In some embodiments, the cytokine domain does not contain a segment that is greater than 80, 85, 90, 95, or 100% identical to amino acids N7-V41 , R1 1 -M37, N102- D144, or Y121 -D144 of SEQ ID NO:1 .
  • the inhibitor binds to IL-1 Rl with K D of less than 100, 50,
  • the inhibitor binds to IL-1 Rl with a better affinity (e.g., lower K D ) and/or a slower dissociation rate than IL-1 ⁇ or IL-1 Ra.
  • the inhibitor does not substantially induce IL-6 expression when contacted to I L- 1 ⁇ responsive human cells.
  • the inhibitor inhibits signaling by IL-1 ⁇ , e.g., with an IC50 of less than 100, 50, 20, 10, or 5 nM.
  • the cytokine domain includes one, two, three or more of the following sequences: WDVNQKTFYLRNNQLVAGYLQGPNV (SEQ ID NO:9);
  • NLEEK (SEQ ID NO:10); RIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEK(SEQ ID NO:1 1 ); AMEADQP(SEQ ID NO:12); FLCTAMEADQPVSLTNMPDEGVMVTKFY(SEQ ID NO:13); and/or sequences at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to the foregoing.
  • the cytokine domain includes one, two, three or more of the following sequences: VQGEESNDKI (SEQ ID NO:14); KKKMEKRF (SEQ ID NO:15); and FSMSFVQGEESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKN YPKKKMEKRFVFNKIEINNKLEFES (SEQ ID NO:16); and/or sequences at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to the foregoing.
  • the inhibitor can include other features described herein.
  • the disclosure provides an isolated protein that includes an amino acid sequence at least 80, 82, 85, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% or 100% identical to a sequence disclosed herein, e.g., a sequence listed in Table 4 or Example 1 , e.g., the amino acid sequence of P01 , P02, P03, P04, P05, P06, or P07, or a sequence in Example 1 , 5, 6, or elsewhere herein.
  • the amino acid sequence includes at least one substitution, insertion, or deletion.
  • the amino acid sequence can include fewer than 15, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, or 2 non- conservative substitutions or fewer than 15, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, or 2 total substitutions.
  • the amino acid sequence can include at least 1 , 2, 3, 4, or 5 substitutions, e.g., conservative substitutions.
  • isolated proteins that include a methionine N-terminal to the amino acid sequence of P01 , P02, P03, P04, P05, P06, or P07, or a sequence in Example 1 , 5, 6, or elsewhere herein, and isolated proteins that include the amino acid sequence of P01 , P02, P03, P04, P05, P06, or P07, or a sequence in Example 1 , 5, 6 or elsewhere herein in which the alanine at N-terminus is absent.
  • sequences can include other features disclosed herein.
  • the sequence can further include a tag, such as a hexa-histidine sequence, e.g., N- or C-terminal relative to the IL-1 Rl binding sequence.
  • the sequence can further include a moiety that modifies the stability or pharmacokinetics of the IL-1 Rl binding sequence.
  • the sequence can further include a serum albumin and/or an Fc domain, or one or more domains thereof, e.g., one or more immunoglobulin constant domains or one or more albumin domains.
  • the protein can have other features described herein.
  • the isolated protein consists of or consists essentially of a sequence or chimeric domain disclosed herein.
  • the disclosure provides an isolated protein that includes a domain having a circularly permuted form of a cytokine domain described herein, e.g., a cytokine domain listed in Table 4 and/or a domain that includes SEQ ID NO:3.
  • the protein can further include a heterologous sequence (such as an Fc domain or albumin) at the N- or C-terminus of the permuted form, optionally spaced by a linker.
  • the protein can have other features described herein.
  • compositions that include one or more receptor binding agents described herein (such as a protein that includes a chimeric cytokine domain).
  • the compositions can be ophthalmic pharmaceutical compositions, topical compositions, or compositions for parenteral administration.
  • the disclosure features a method of modulating an immune or inflammatory response in a subject.
  • the method can include administering a
  • composition that includes a receptor binding agent described herein to a subject in an amount effective to modulate the immune or inflammatory response in the subject.
  • the disclosure features a method of treating an IL-1 mediated disorder in a subject.
  • the method includes administering a composition that includes a protein that can bind to IL-1 Rl, e.g., a receptor binding agent described herein, to the subject.
  • the disorder can be an autoimmune disorder, e.g., rheumatoid arthritis or juvenile chronic arthritis, scleroderma, Sjogren's syndrome, ankylosing spondylitis, Behcet's syndrome, an inflammatory bowel disease, asthma, vasculitis, or psoriasis.
  • the disorder can be a disorder associated with aggregate formation, e.g., hyperuricemia, gout, diabetes (including non-insulin dependent diabetes), Alzheimer's disease, secondary reactive amyloidosis, amyotrophic lateral sclerosis (ALS),
  • hyperuricemia e.g., hyperuricemia, gout, diabetes (including non-insulin dependent diabetes), Alzheimer's disease, secondary reactive amyloidosis, amyotrophic lateral sclerosis (ALS),
  • ALS amyotrophic lateral sclerosis
  • the disorder can also be a CAPS (CIAS1 Associated Periodic Syndromes) disorder or other disorder described herein.
  • CAPS CIAS1 Associated Periodic Syndromes
  • the disclosure features a method of treating an IL-1 mediated ocular disorder in a subject.
  • the method can include administering a composition that includes a protein that can bind to IL-1 Rl, e.g., a receptor binding agent described herein, to the subject.
  • the composition is an ophthalmic composition that is administered topically to an eye of the subject or surrounding region.
  • the disorder is a dry eye disorder.
  • the subject does not exhibit manifestations of systemic autoimmune disease.
  • the subject has Sjogren's syndrome.
  • the subject has graft-versus- host disease (GVHD).
  • the disorder is uveitis.
  • the disclosure features a method of inhibiting IL-1 activity.
  • the method includes contacting a receptor binding agent that can bind to IL-1 Rl to cells responsive to IL-1 or to a subject.
  • the protein is provided in an amount effective to inhibit IL-1 activity associated with the cells or in the subject.
  • the protein can be contacted to cells from a subject ex vivo.
  • this disclosure features an isolated nucleic acid that includes one or more sequences encoding the proteins described herein or a nucleic acid disclosed herein (e.g., in Table 5), a sequence that hybridizes to such nucleic acid, or that is at 80, 82, 85, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% or 100% identical to such nucleic acid.
  • Exemplary hybridizing sequences can be at least 200, 300, 400, 420, or 450 nucleotides in length, e.g., between 420-480 nucleotides in length.
  • the nucleic acid can also include other features disclosed herein.
  • a receptor binding agent can be produced by a method that includes maintaining the host cell under conditions that permit expression of the receptor binding agent, and optionally recovering the receptor binding agent, e.g., from cells or media associated with the host cell.
  • the receptor binding agent can be purified from lysate from the cells.
  • the purified receptor binding agent can be formulated, e.g., with one or more of an excipient, a stabilizer, and a buffer.
  • the method includes identifying at least two parental proteins having a common fold (e.g., a first parental protein and a second parental protein), locating at least two segments within the first parental protein and constructing a nucleic acid that has a sequence encoding a chimeric amino acid sequence that includes the two segments from the first parental protein and residues that are predominantly from the second parental protein at remaining positions.
  • the domain can be a domain that is largely composed of ⁇ -sheets, or a domain that is largely composed of a-helices, or a domain that has a combination of such elements.
  • the domain can have the fold of a cytokine.
  • the first and second parental proteins can be related by homology, e.g., between 10-40% amino acid identity.
  • the segments from the first protein are located within a single folded protein domain, and the chimeric amino acid sequence includes a form of the folded protein domain that is non-identical to the corresponding domain in the first and second parental protein.
  • the two parental protein have different functional properties, and the chimeric domain can have properties of one or both of the parental proteins.
  • the chimeric domain has one binding interface from the first parental protein, and another binding interface from the second parental protein.
  • Site A residues in IL-1 ⁇ include: ARG1 1 , SER13, GLN14, GLN15,
  • Site A residues residues of other IL1 cytokine family members
  • site A residues residues of other IL1 cytokine family members
  • Site A region as shown in Fig.
  • Site B Site B residues in IL-1 ⁇ include: ALA1 , PR02, ARG4, GLN48, GLU51 , ASN53, ILE56, LYS92, LYS93, LYS94, LYS103, GLU105, and ASN108, and
  • Site B residues residues of other IL1 cytokine family members.
  • site B residues residues of other IL1 cytokine family members
  • SER152 which are outside the Site B region in Fig. 4.
  • a "Site B region" as shown in Fig. 4, including for example a B1 segment (corresponding in IL-1 ⁇ to 1 -5 of SEQ ID NO:1 ), a B2 segment (corresponding in IL-1 ⁇ to 48-56 of SEQ ID NO:1 ), and a B3 segment (corresponding in IL-1 ⁇ to 92-98 of SEQ ID NO:1 ), and corresponding segments in other IL1 cytokine family members.
  • Site C residues in IL-1 ⁇ include: ILE104, ILE106, ASN107, LYS109, GLU1 1 1 , THR137, LYS138, GLY139, GLY140, GLN141 , THR144, and ASP145 and corresponding residues of other IL1 cytokine family members (referred to herein as "Site C residues").
  • Site C residues it is possible to define a "Site C region" as shown in Fig. 4, including for example a C1 segment (corresponding in IL-1 ⁇ to 136-145 of SEQ ID NO:1 ), and corresponding segments in other IL1 cytokine family members.
  • Site D residues in IL-1 ⁇ include: LEU6, THR9, LYS63, GLU64, LYS65, and ASN66 and corresponding residues of other IL1 cytokine family members (referred to herein as "Site D residues").
  • Site D residues it is possible to define a "Site D region" as shown in Fig. 4, including, for example, a D1 segment (corresponding in IL-1 ⁇ to 6-9 of SEQ ID NO:1 ), a D2 segment (corresponding in IL-1 ⁇ to 37-41 of SEQ ID NO:1 ), and a D3 segment (corresponding in IL-1 ⁇ to 63-66 of SEQ ID NO:1 ), a D4 segment
  • amino acid sequence of IL-1 ⁇ (human) as referenced herein is:
  • IL-1 Ra human
  • IL-1 ⁇ , IL-1 a, and IL-1 Ra refer to the respective mature proteins.
  • amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid "homology”).
  • the percent identity between the two sequences is a function of the number of identical positions shared by the sequences. To determine collective identity of one sequence of interest to a group of reference sequences, a position is considered to be identical if it is identical to at least one amino acid at a corresponding position in any one or more of the group of reference
  • identity can be calculated collectively for all members of such list to arrive an overall percentage identity.
  • sequences that are at least 80, 82, 85, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, or 99% identical to sequences disclosed herein.
  • corresponding to is used to designate the position of an amino acid residue in a polypeptide of interest with respect to a reference
  • polypeptide In general the position is the one indicated by an alignment provided herein (e.g., Fig. 4).
  • hybridizes under high stringency conditions describes conditions for hybridization and washing.
  • Guidance for performing hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N. Y. (1989), 6.3.1 -6.3.6, which is incorporated by reference. Aqueous and nonaqueous methods are described in that reference and either can be used.
  • High stringency hybridization conditions include hybridization in 6X SSC at about 45 °C, followed by one or more washes in 0.2X SSC, 0.1 % SDS at 65 ⁇ , or substantially similar conditions.
  • isolated nucleic acids that contain sequences that hybridize under high stringency conditions to nucleic acids encoding amino acid sequences disclosed herein and to the nucleic acids disclosed herein, e.g., in Example 1 .
  • Naturally occurring proteins referenced herein specifically include the human form of such protein, and also forms from other mammalian species. All patents, published patent applications, and published references cited herein are incorporated by reference for all purposes.
  • FIG. 1 is a graphic of the structure of P04 as determined from X-ray
  • FIG. 2 depicts three views of a model of the P05 protein bound to the extracellular domain of human IL-1 Rl.
  • IL-1 Ra residues are depicted in black and IL-1 ⁇ residues are depicted in white
  • FIG. 3 depicts models of chimeric proteins in which IL-1 Ra residues are depicted in black and IL-1 ⁇ residues are depicted in white.
  • the model depicts the proteins: P01 (FIG. 3A), P03 (FIG. 3B), P04 (FIG. 3C), P05 (FIG. 3D), P07 (FIG. 3E), and P06 (FIG. 3F).
  • FIG. 4 provides an alignment of several human IL-1 family cytokines: IL-1 ⁇ (SEQ ID NO:1 ), IL-1 oc (SEQ ID NO:2), IL-1 Ra (SEQ ID NO:3), IL-33 (SEQ ID NO:4), IL-36Ra (SEQ ID NO:5), IL-36oc (SEQ ID NO:6), ⁇ -36 ⁇ (SEQ ID NO:7), and IL-36y (SEQ ID NO:8). Segments referenced in the text herein are identified under the alignment. In addition, ⁇ -sheets and loops between such sheets are identified.
  • FIG. 5A is a listing of the amino acid sequence of P01 (SEQ ID NO:17).
  • FIG. 5B is a listing of the amino acid sequence of P02 (SEQ ID NO:18).
  • FIG. 5C is a listing of the amino acid sequence of P03 (SEQ ID NO:19).
  • FIG. 5D is a listing of the amino acid sequence of P04 (SEQ ID NO:20).
  • FIG. 5E is a listing of the amino acid sequence of P05 (SEQ ID NO:21 ). Segments from IL-1 ⁇ are shown in bold italics. See also
  • FIG. 6 is an image of an SDS-PAGE gel showing exemplary samples of protein purified from E. coli cells expressing receptor binding agents. The 15 and 20 kDa molecular weight markers are indicated at left. Lanes are as follows: molecular weight marker (lane 1 and 6), extract (lanes 2 and 7), material purified by cation exchange chromatography (lanes 3 and 8), material additionally purified by anion exchange chromatography (lanes 4 and 9), and reduced samples of such material (lanes 5 and 10). Lanes 2-5 are of P05 purification, and Lanes 6-10 are of P04 purification. See also Example 2. FIG.
  • FIG. 7 A is a table and accompanying bar graph showing the ability of the P06, P07, and P01 proteins to agonize signaling relative to IL-1 ⁇ and a negative control, ⁇ - glucuronidase (GUS) protein.
  • FIG. 7B is a graph depicting antagonism of IL-1 ⁇ at various I L-1 ⁇ concentrations by P01 .
  • FIG. 8A is a graph depicting IL-1 ⁇ antagonism by P03 (hexa-histidine tagged),
  • FIG. 8B is a graph depicting IL-1 ⁇ antagonism by lysates containing untagged forms of P01 , P02, P03, P04, and P05, and IL-1 Ra in the presence of 0.1 ng/ml IL-1 ⁇ (human) and using estimates of the concentration of protein in the respective lysates.
  • FIG. 9 contains graphs of SPR data showing binding kinetics to immobilized soluble IL-1 Rl for the following proteins: IL-1 ⁇ (FIG. 9A), IL-1 Ra (FIG. 9B), P04 (FIG. 9C), and P05 (FIG. 9D).
  • FIG. 1 0A is a graph depicting thermal denaturation of IL-1 Ra, IL-1 ⁇ , P03, P04, and P05 as described in Example 7.
  • FIG. 10B depicts the negative first derivative of the graph in FIG. 10A.
  • FIG. 1 1 A is a bar graph showing the mean corneal staining score ⁇ SEM as tested by fluorescein staining of the cornea per eye of two independent studies, on days 0, 3, 7, 9, and 1 1 for mice in a dry eye model.
  • Asterisks indicate statistical significance of P05 relative to vehicle as follows: * (P ⁇ 0.05) and ** (P ⁇ 0.005).
  • FIG. 1 1 B is a bar graph representing data from a separate experiment showing mean corneal staining score ⁇ SEM of the cornea per eye, on days 0, 3, 7, 9, and 1 1 for mice in a dry eye model.
  • Asterisks indicate statistical significance of P05 relative to murine serum albumin as follows: * (P ⁇ 0.05) and *** (P ⁇ 0.0005).
  • Asterisks indicate statistical significance of P05 relative to Restasis® as follows: ** (P ⁇ 0.005) and *** (P ⁇ 0.0005).
  • FIG. 12A depicts the structure of the X-ray crystallographic structure (black) of P04 overlaid on a computed model (gray) of its structure.
  • FIG. 12B illustrates interactions between K64 and E39 of P04;
  • FIG. 12C illustrates interactions between the C-terminal residues Q149 and S152 with K40 and R9 of P04.
  • the IL-1 family of cytokines includes several members, all having a common ⁇ - trefoil fold comprised of six ⁇ -strands that form a ⁇ -barrel capped by another six ⁇ - strands.
  • the primary structures of the human and other mammalian forms of these cytokines are known.
  • An exemplary structural alignment of several human IL-1 family members is shown in Fig. 1 .
  • IL-1 fold is highly plastic.
  • elements from different members can be combined to provide proteins that agonize or antagonize cytokine signaling.
  • these proteins include chimeric cytokine domains that include, for example, two or more segments or surface residues from one cytokine in the context of another cytokine or a cytokine consensus sequence, thus creating non-naturally occurring combinations of receptor interaction sites from different IL-1 family cytokines.
  • IL-1 family cytokines can include at least two primary receptor interaction sites, referred to as Site A and Site B.
  • Sites A and B are involved in contacts to the primary cytokine receptor (e.g., IL-1 Rl).
  • the primary cytokine receptor e.g., IL-1 Rl
  • the two proteins differ substantially with respect to sites A and B such that IL-1 Ra makes fewer receptor contacts in Site B than Site A.
  • chimeric cytokine domains that include a Site A derived from one cytokine and a Site B derived from another cytokine.
  • the plasticity of the IL-1 fold permits construction of a variety of chimeric cytokine domains to antagonize IL-1 signaling.
  • IL-1 family cytokines can include two secondary receptor interaction sites, referred to as Site C and Site D, which are involved in agonism and/or antagonism, and can be determinative of a cytokine's ability to interact with its secondary cytokine receptor (e.g., IL-1 RAcP).
  • Site C and Site D residues from natural receptor antagonists can impart antagonistic properties.
  • Chimeric cytokine domains can be constructed that include one or both of Sites A and B from one or more IL-1 agonists (such as IL-1 ⁇ and IL-1 a) and/or an IL-1 receptor antagonist (such as IL-1 Ra) and one or both of Sites C and D from an IL-1 receptor antagonist (such as IL-1 Ra). Accordingly, it is possible to produce a chimeric cytokine domain that antagonizes signaling and that includes Site B residues from an IL-1 agonist.
  • IL-1 agonists such as IL-1 ⁇ and IL-1 a
  • an IL-1 receptor antagonist such as IL-1 Ra
  • the source sequences can be identical to the human sequences for the identified cytokine domains or can contain mutations relative to the human sequences, e.g., such that they are at least 70%, 75%, 80%, 85%, 90%, 95% or more identical to the human sequences in each respective region, e.g., they can include one or more segments from each region.
  • Sources for Site A residues and Site B residues can be chosen to maximize affinity for the primary receptor.
  • Site A residues can be derived from IL-1 Ra
  • Site B residues can be derived from IL-1 ⁇ .
  • a chimeric cytokine domain can have the ability to bind an IL-1 family receptor, e.g., human IL-1 Rl with a K D of less than 10 "8 , 10 "9 , or 10 "10 , e.g., a K D within 10 or 100 fold that of a natural receptor ligand (e.g., I L- 1 ⁇ , IL-1 a, or IL-1 Ra) under the same conditions or less than that of a natural ligand (e.g., I L- 1 ⁇ , IL-1 a, or IL-1 Ra) under the same conditions.
  • the chimeric cytokine domain binds with a K D less than, a K on faster than, or a K off slower than at least one of its parental cytokine domains.
  • Chimeric cytokine domains that bind to IL-1 family receptor and which antagonize receptor signaling can be used as receptor binding agents, e.g., to treat disorders mediated by IL-1 family cytokine signaling as described below.
  • the chimeric cytokine domain binds to IL-1 Rl and antagonizes IL-1 signaling.
  • it has an IC 50 of less than 100, 10, 1 , 0.6, or 0.3 nM.
  • the cytokine domain can be at least 40, 45, or 50% identical, but less than completely identical, e.g., less than 95, 90, 85, or 80% identical to a first IL-1 family cytokine domain.
  • the cytokine domain can also be at least 40, 45, or 50% identical, but less than completely identical, e.g., less than 95, 90, 85, or 80% identical to a second IL-1 family cytokine domain.
  • the first and second IL-1 family cytokine domain can be less than 50% identical to each other.
  • the first IL-1 family cytokine domain can be an agonist (e.g., IL-1 ⁇ or IL-1 a), whereas the second IL-1 family cytokine domain can be a receptor antagonist (e.g., IL-1 Ra).
  • At least 80, 85, 90, 92, 94, 95, 97, 98, 99, or 100% of the positions within the cytokine domain have the property that, at each such position, the amino acid present is either identical to the first IL-1 family cytokine domain or to the second IL-1 family cytokine domain (or both if the first and second IL-1 family cytokine are identical at the particular position). Where 100% of the amino acid positions in the cytokine domain have this property, the domain is a complete chimera of two cytokines.
  • Chimeric cytokine domains can also be made from more than two cytokines and can also have mutations relative to its parental cytokines (e.g., one or more particular positions where the amino acid present differs from the corresponding amino acid in each of its parental cytokines).
  • Cytokine domains can have Site A, B, C, and D residues from different IL-1 cytokine domains, and likewise can have Site A, B, C, and D regions from different IL-1 cytokine domains.
  • cytokine domain For example, in certain embodiments, a cytokine domain
  • a receptor antagonist e.g., IL-1 Ra
  • an agonist at at least 5, 10, 12, 15, 16, 17, or 18 of the Site A residues identified above, or at at least 5, 10, 12, 15, 16, 17, 18, 19, or 20 of the extended Site A residues identified above, or
  • the cytokine domain includes residues that are at least 70, 75, 80, 85, 88, 90, 92, 95, or 100% identical to segments A1 , A2, or A1 +A2 in a receptor antagonist (e.g., IL-1 Ra) or an agonist (e.g., IL-1 ⁇ or IL-1 a).
  • a receptor antagonist e.g., IL-1 Ra
  • an agonist e.g., IL-1 ⁇ or IL-1 a
  • a cytokine domain includes residues identical to an IL-1 agonist (e.g., IL-1 ⁇ residues) at at least 5, 10, 12, 15, 16, 17, or 18 of the Site A residues identified herein, or at least 5, 10, 12, 15, 16, 17, or 18 of the extended Site A residues identified above, or conservative substitutions of such residues, or at least 50, 65, 75, 80, 90, 95, or 100% of such residues.
  • an IL-1 agonist e.g., IL-1 ⁇ residues
  • a cytokine domain includes residues identical to an IL-1 agonist (e.g., IL-1 ⁇ or IL-1 a residues) at at least 2, 3, 5, 8, 9, 10, 1 1 , 12, 13, 14, or 15 of the Site B residues identified herein, or conservative substitutions of such residues, or at least 50, 65, 75, 80, 90, 95, or 100% of such residues.
  • an IL-1 agonist e.g., IL-1 ⁇ or IL-1 a residues
  • the cytokine domain includes residues that are at least 70, 75, 80, 85, 88, 90, 92, 95, or 100% identical to segments B1 , B2, B3, B1 +B2, B1 +B3, B2+B3, or B1 +B2+B3 in an IL-1 cytokine agonist (e.g., IL-1 ⁇ or ll_-1 oc).
  • an IL-1 cytokine agonist e.g., IL-1 ⁇ or ll_-1 oc.
  • a cytokine domain includes residues identical to a receptor antagonist (e.g., IL-1 Ra residues) at at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 of the Site C residues identified herein, or conservative substitutions of such residues, or at least 50, 65, 75, 80, 90, or 100% of such residues.
  • the cytokine domain includes residues that are at least 50, 65, 75, 80, 90, or 100% identical to the segment C1 in a receptor antagonist (e.g., IL-1 Ra).
  • the cytokine domain can include, for example, one or more of: a hydrophobic amino acid (e.g., Met or lie) at the position corresponding to THR137 of SEQ ID NO:1 , or a hydrophobic, e.g., an aliphatic amino acid (e.g., Val or lie), at the position corresponding to GLN141 of SEQ ID NO:1 , and a non-acidic amino acid, such as a basic amino acid (e.g., Lys or Arg) at the position corresponding to
  • a non- acidic amino acid such as a basic amino acid (e.g., Lys or Arg) or a hydrophobic amino acid, is located at the position corresponding to ASP145 of SEQ ID NO:1 .
  • a cytokine domain includes residues identical to a receptor antagonist (e.g., IL-1 Ra residues) at at least one, two, three, four, five, or six of the Site D residues identified herein, or a conservative substitution of such residue, or at least 50, 65, 75, 80, 90, 95, or 100% of such residues.
  • the cytokine domain includes residues that are at least 50, 65, 75, 80, 90, 95, or 100% identical to segments D1 , D2, D3, D4, D5, D1 +D2, D1 +D2+D3, and combinations thereof in a receptor antagonist (e.g., IL-1 Ra).
  • residues in IL-1 ⁇ contact or are in proximity with IL-1 Rl for example: ALA1 , PR02, VAL3, ARG4, LEU6, ARG1 1 , SER13, GLN14, GLN15, GLU25, LYS27, LEU29, HIS30, LEU31 , GLN32, GLY33, GLN34, ASP35, MET36, GLN38, GLN39, PHE46, GLN48, GLU51 , SER52, ASN53, LYS55, ILE56, PR057, LYS92, LYS93, LYS94, LYS103, GLU105, ASN108, ALA127, GLU128, ASN129, MET130, GLN141 , GLN149, PHE150, and SER152.
  • these residues can be classified into two sets: Set 1 and Set 2.
  • Exemplary Set 1 residues in IL-1 ⁇ include: ARG1 1 , SER13, GLN14, GLN15, GLU25, LYS27, LEU29, HIS30, LEU31 , GLN32, GLY33, GLN34, ASP35, MET36, GLN38, GLN39, ALA127, GLU128, ASN129, MET130, and GLN141 and corresponding residues in other IL-1 cytokine family members.
  • Extended Set 1 residues include interaction Set 1 residues and residues within 4 Angstroms of the foregoing in the 1 1TB structure.
  • Exemplary Set 2 residues in IL-1 ⁇ include: ALA1 , PR02, VAL3, ARG4, LEU6, PHE46, GLN48, GLU51 , SER52, ASN53, LYS55, ILE56, PR057, LYS92, LYS93, LYS94, LYS103, GLU105, ASN108, GLN149, PHE150, and SER152 and corresponding residues in other IL-1 cytokine family members.
  • Extended Set 2 residues include interaction Set 2 residues and residues within 4 Angstroms of the foregoing in the 1 1TB structure.
  • a cytokine domain includes IL-1 ⁇ residues at at least 15, 16, 17, 18, 19, 20, or 21 of the Set 1 residues identified above.
  • a cytokine domain includes IL-1 ⁇ residues at at least 15, 16, 17, 18, 19, 20, 21 , or 22 of the Set 2 residues identified above. In some embodiments, a cytokine domain includes IL-1 ⁇ residues at at least 15, 16, 17, 18, 19, 20, or 21 of the extended Set 1 residues. In some embodiments, a cytokine domain includes IL-1 ⁇ residues at at least 15, 16, 17, 18, 19, 20, or 21 of the extended Set 2 residues.
  • variants that can be used as a receptor binding agent include proteins having sequences derived from two or more IL-1 cytokine family members. Examples of such variants include chimeric domains based on IL-1 ⁇ and IL-1 Ra.
  • the variants can include one or more amino acid residues from Set 1 of IL-1 Ra (e.g., all Set 1 residues from IL-1 Ra) and one or more amino acid residues from Set 2 of IL-1 ⁇ (e.g., all Set 2 residues from IL-1 ⁇ ).
  • Exemplary chimeric proteins are predominantly (e.g., at least 50, 60, 70, 75, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 97, 98, 99, or 100%) identical to IL-1 ⁇ at the following amino acids positions (i.e., based on correspondence to these positions in IL-1 ⁇ ):
  • residues 1 -8, 42-120, and 141 -153 of SEQ ID NO:1 residues 1 -6, 45-61 , 86-95, and 148-153 of SEQ ID NO:1 ; and residues 1 -10, 37-125, and 131 -153 of SEQ ID NO:1 .
  • the remaining residues can be predominantly (e.g., at least 50, 60, 70, 75, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 97, 98, 99, or 100%) identical to IL-1 Ra.
  • the following amino acid positions can be predominantly identical to IL-1 Ra: residues 9-41 and 121 -140 of SEQ ID NO:1 ; residues 7-44, 62-85, and 96-147 of SEQ ID N01 ; and residues 1 1 -36 and 126-130 of SEQ ID NO:1 .
  • the cytokine domain is identical to IL-1 ⁇ at at least 2, 4, 5, 10, or 20 positions in addition to amino acid positions Gln48 - Asn53 of IL-1 ⁇ , and, e.g., the cytokine domain is predominantly, e.g., at least 50, 60, 70, 75, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 97, 98, 99, or 100% identical to a cytokine other than I L- 1 ⁇ .
  • the cytokine domain includes at least 3, 4, 5, 6, 7, or 8 residues identical to IL-1 ⁇ at positions corresponding to 1 -8 of SEQ ID NO:1 .
  • it includes residues identical to IL-1 ⁇ at positions corresponding to 3, 4, or all 5 of: ALA1 , PR02, VAL3, ARG4, and LEU6.
  • the cytokine domain includes at least 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, or 17 residues identical to IL-1 ⁇ at positions corresponding to 45-61 of SEQ ID NO:1 .
  • it includes residues identical to IL-1 ⁇ at positions corresponding to 3, 4, 5, 6, 7 or 8 of: PHE46, GLN48, GLU51 , SER52, ASN53, LYS55, ILE56, and PR057.
  • the cytokine domain includes at least 5, 6, 7, 8, 9, or 10 residues identical to IL-1 ⁇ at positions corresponding to 86-95 of SEQ ID NO:1 .
  • it includes residues identical to IL-1 ⁇ at positions corresponding to one, two, or all three of LYS92, LYS93, and LYS94.
  • the cytokine domain includes at least 3, 4, 5, or 6 residues identical to IL-1 ⁇ at positions corresponding to 148-153 of SEQ ID NO:1 .
  • it includes residues identical to IL-1 ⁇ at positions corresponding to one, two, or all three of GLN149, PHE150, and SER152.
  • the cytokine domain does not include a threonine at the position corresponding to THR147 in IL-1 ⁇ .
  • this position can be an aromatic, such as a tyrosine.
  • the aromatic at the position corresponding to THR147 in IL-1 ⁇ can pack against another aromatic (e.g., tryptophan) present in several embodiments at position 1 1 (according to IL-1 ⁇ numbering).
  • the cytokine domain does not include an aromatic at the position corresponding to CYS8 in IL-1 ⁇ .
  • this position can be an amino acid other than phenylalanine, or other than an aromatic.
  • it can be cysteine, valine, serine, threonine or alanine.
  • This position is located near other bulky residues, particularly MET44 and MET148 (according to I L-1 ⁇ numbering).
  • MET44 and MET148 are bulky residues.
  • the cytokine domain includes at least five, six, or seven residues identical to IL-1 Ra at positions corresponding to 30-36 of SEQ ID NO:1 , e.g., at least five, six, or seven residues identical to YLQGPNV (SEQ ID NO:43).
  • the cytokine domain includes at least 10, 12, 14, 16, 18, 19, 20, 21 , 22, 23, 24, 25, or 26 residues identical to IL-1 Ra at positions corresponding to 1 1 -36 of SEQ ID NO:1 .
  • the cytokine domain can also include a basic residue at the position corresponding to 145 of SEQ ID NO:1 .
  • the cytokine domain includes: (a) glutamic acid at the position corresponding to Val40 of IL-1 ⁇ and lysine at the position corresponding to Lys65 of IL-1 ⁇ ; (b) arginine at the position corresponding to Thr9 of IL-1 ⁇ and glutamine at the position corresponding to Gln149 of IL-1 ⁇ ; and/or (c) lysine at the position corresponding to V41 of IL-1 ⁇ and serine at the position corresponding to Ser152 of IL-1 ⁇ .
  • the cytokine domain can also include at least four, five, six, or seven residues identical to IL-1 Ra at positions corresponding to 126-132, e.g., at least four, five, six, or seven residues identical to MEADQPVS (SEQ ID NO:44).
  • the cytokine domain includes at least 10, 12, 14, 16, 18, 19, 20, 21 , 22, 23, or 24 residues identical to IL-1 ⁇ at positions corresponding to 62-85 of SEQ ID NO:1 .
  • the cytokine domain includes one or more of the following amino acid sequences (e.g., all four of): RSLAFR (SEQ ID NO:45), IDVSFV (SEQ ID NO:46), KKMDKR (SEQ ID NO:47), and KFYMQF (SEQ ID NO:48); one or more of the following (e.g., all four of) RSLAFR (SEQ ID NO:45), IDVSFV (SEQ ID NO:46), NKLSFE (SEQ ID NO:49), and KFYMQF (SEQ ID NO:48); one or more of the following (e.g., all four of) RSLAFR (SEQ ID NO:45), EEKFSM (SEQ ID NO:50), RFVFIR (SEQ ID NO:51 ), and VTKFTM (SEQ ID NO:52); one or more of the following (e.g., all four of) RSLAFR (SEQ ID NO:45), EEKFSM (SEQ ID NO:50
  • Chimeric proteins can be used for a variety of purposes. For example, they can be used to increase or decrease receptor signaling activity, to detect cells expressing receptors, or to purify cells or proteins to which they bind.
  • Specific examples of chimeric cytokine domains that are based on IL-1 ⁇ and IL-Ra are provided in Example 1 below and include the following exemplary cytokine domains that antagonize IL-1 signaling:
  • the P01 domain includes three segments from IL-1 Ra corresponding to amino acids Ala12-Val48, Ne60-Val83, and Asp95-Tyr147 of SEQ ID NO:3, and the remaining four segments from IL-1 ⁇ . Overall, the P01 domain has 74 of 153 amino acids from IL-1 ⁇ (about 48% identity) and 1 19 amino acids from IL-1 Ra (about 77% identity). These percentages add up to greater than 100% because a number of amino acids in P01 and other exemplary proteins disclosed herein are amino acids that are conserved between IL-1 ⁇ and IL-1 Ra and accordingly contribute to the percentage identity for both IL-1 ⁇ and IL-1 Ra.
  • the P02 domain includes three segments from IL-1 Ra corresponding to amino acids Ala12-Val48, Ile60-Val83, and Ser1 10-Tyr147 of SEQ ID NO:3, and the remaining four segments from IL-1 ⁇ . Overall, the P02 domain has 85 of 153 amino acids from IL-1 ⁇ (about 55% identity) and 108 amino acids from IL-1 Ra (about 70% identity).
  • the P03 domain includes two segments from IL-1 Ra corresponding to amino acids Ala12-Lys45 and Phe100-Lys145 of SEQ ID NO:3, and the remaining three segments from IL-1 ⁇ . Overall, the P03 domain has 94 of 153 amino acids from I L- 1 ⁇ (about 61 % identity) and 91 amino acids from IL-1 Ra (about 64% identity).
  • the P04 domain includes two segments from IL-1 Ra corresponding to amino acids Ala12-Lys45 and Ala1 14-Lys145 of SEQ ID NO:3, and the remaining three segments from IL-1 ⁇ . Overall, the P04 domain has 104 of 153 amino acids from IL-1 ⁇ (about 68% identity) and 89 amino acids from IL-1 Ra (about 58% identity).
  • the P05 domain includes two segments from IL-1 Ra corresponding to amino acids Arg14-Lys45 and Phe120-Tyr147 of SEQ ID NO:3, and the remaining three segments from IL-1 ⁇ . Overall, the P05 domain has 108 of 153 amino acids from IL-1 ⁇ (about 70% identity) and 85 amino acids from IL-1 Ra (about 55% identity).
  • chimeric proteins can be similarly constructed.
  • chimeric proteins can be made between IL-1 a and IL-1 Ra, e.g., specifically domains including the identified segments of IL-1 Ra in combination with corresponding residues from IL-1 a rather than IL-1 ⁇ for each of the above examples.
  • Protein sequences such as those described herein, can be varied, e.g., by making one or more conservative substitutions.
  • Conservative substitutions can be made to retain function or to make modest changes in function.
  • Exemplary conservative substitutions are described in the following table:
  • Substitutions can be chosen based on their potential effect on (a) backbone structure in the vicinity of the substitution, for example, a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the volume and branching of the side chain.
  • Amino acid residues can be classified based on side-chain properties: (1 ) hydrophobic: norleucine, met, ala, val, leu, ile;(2) neutral hydrophilic: cys, ser, thr; asn; gin; (3) acidic: asp, glu; (4) basic: his, lys, arg; (5) residues that affect backbone conformation: gly, pro; and (6) aromatic: trp, tyr, phe.
  • Non-conservative substitutions can include substituting a member of one of these classes for a member of a different class.
  • Conservative substitutions can include substituting a member of one of these classes for another member of the same class.
  • the significance of a particular residue can also be evaluated in the context of the hydropathic index for the amino acid. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics:
  • tryptophan (-0.9); tyrosine (-1 .3); proline (-1 .6); histidine (-3.2); glutamate (-3.5);
  • glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5).
  • hydropathic amino acid index and its significance see, for example, Kyte et al., 1982, J. Mol. Biol. 157:105-131 .
  • sequence of a protein can be varied by any method, including oligonucleo- tide-mediated (site-directed) mutagenesis, (Carter et al., Nucl. Acids Res. ,13:4331 , 1986; Zoller et al., Nucl. Acids Res., 10:6487, 1987), cassette mutagenesis (Wells et al., Gene, 34:315, 1985), restriction selection mutagenesis (Wells et al., Philos. Trans. R. Soc. London, 317:415, 1986), and PCR mutagenesis.
  • oligonucleo- tide-mediated (site-directed) mutagenesis (Carter et al., Nucl. Acids Res. ,13:4331 , 1986; Zoller et al., Nucl. Acids Res., 10:6487, 1987), cassette mutagenesis (Wells et al., Gene, 34:315, 1985), restriction selection mutagenesis (Well
  • Scanning amino acid analysis can be employed to evaluate one or more amino acids along a contiguous sequence.
  • the method can involve mutating each or nearly each amino acid in a region to a particular amino acid, e.g., to a relatively small, neutral amino acid such as alanine, serine, or valine.
  • Alanine is typically chosen because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main- chain conformation of the variant (Cunningham and Wells, Science, 244: 1081 -1085, 1989). It is also the most common amino acid and is frequently found in both buried and exposed positions (Creighton, The Proteins, (W. H. Freeman & Co., N.Y.); Chothia, J. Mol.
  • the scanning process can also be adapted to make more marked changes, e.g., charged residues can be changed to residues of the opposite charge, residues with short side chains can be replaced with ones with bulk side chains.
  • arginine scanning is an approach that can be used instead of or in addition to alanine scanning.
  • the scanning can be applied to each residue in a region or to residues of a particular property, e.g., residues at or near the surface of a protein or likely to be at or near the surface of the protein.
  • the structure of a protein, one of its domains, or a complex involving the protein can be modeled, e.g., by performing homology based modeling, energy minimization and/or other modeling using known solved structures.
  • Such methods include: Accelrys Software Inc., Discovery Studio®, Release 3.0 , San Diego: Accelrys Software Inc., 2010, AMBERTM modeling software (Case et al. (2005) J. Computat. Chem. 26, 1668- 1688 and Case et al. (2010), AMBER 1 1 , University of California, San Francisco, CA USA) and CHARMMTM modeling software (Molecular Simulations Inc.). See also generally Baker and Sali, Science 294(5540):93-6, 2001 ).
  • the structure can also be determined directly, e.g., using X-ray crystallography and/or NMR spectroscopy.
  • Exemplary PDB structures describing the structure of IL-1 family cytokines include: 1 11 B, 1 1LR, 1 1RA, 1 1TB, 211 B, 2ILA, 2KLL, 411 B, 511 B, 611 B, 711 B, 811 B, 9ILB, and 1 MD6 (available by http from www.pdb.org from RCSB-Rutgers, Piscataway NJ, USA, and from the National Library of Medicine (Bethesda, MD, USA)).
  • the structures of IL-1 ⁇ alone and in complex with the receptor IL-1 Rl have been solved. See e.g., Finzel et al. (1989) J. Mol. Biol.
  • Homology modeling can be assisted by alignment of sequences, e.g., using computer software such as Basic Local Alignment Search Tool (BLAST), PSI-BLAST, PHI-BLAST, WU-BLAST-2, and/or MEGABLAST.
  • BLAST Basic Local Alignment Search Tool
  • PSI-BLAST PSI-BLAST
  • PHI-BLAST PHI-BLAST
  • WU-BLAST-2 WU-BLAST-2
  • MEGABLAST Basic Local Alignment Search Tool
  • Additional algorithms for aligning macromolecules include FASTA (Pearson, 1995, Protein Science 4, 1 145-1 160), ClustalW (Higgin et al., 1996, Methods Enzymol. 266, 383-402), DbClustal (Thompson et al., 2000, Nucl. Acids Res. 28, 2910-2926), and the Molecular Operating Environment (Chemical Computing Group, Montreal, Quebec Canada H3A 2R7).
  • FASTA Protein Science 4, 1 145-1 160
  • ClustalW Higgin et al., 1996, Methods Enzymol. 266, 383-402
  • DbClustal Thompson et al., 2000, Nucl. Acids Res. 28, 2910-2926
  • the algorithm of Myers and Miller Myers & Miller,
  • CABIOS 4, 1 1 -17, 1988 which is incorporated into the ALIGN program (version 2.0) of the GCG sequence alignment software package can be used.
  • a consensus sequence for IL-1 ⁇ and IL-1 Ra can be obtained by comparing the two sequences and identifying residues that are identical or that are highly conserved.
  • a chimeric cytokine domain described herein can have at least 60, 70, 80, 90, 95, or 100% of such identical residues or highly conserved residues.
  • An exemplary consensus sequences is: DXXQKX ⁇ 8 _ 9 ⁇ L-AXXLQGX ⁇ 18 - 28 ⁇ LGX ⁇ 7 ⁇ LSCVXXXDXXLQLEXVX ⁇ 8 _ 9 ⁇ KXXKRFXFX ⁇ 10 ⁇ FESAXXPXWXXXTXXXXXXPVXLX ⁇ 5 - 6 ⁇ GXXXTXFXXQ (SEQ ID NO:57), where X is independently any amino acid and the subscripted number or range is the number of occurrences.
  • a chimeric cytokine domain described herein can have at least 60, 70, 80, 90, 95, or 100% identity to the consensus sequence (wherein X residues are not counted towards identity). Other consensus sequences can be identified in like manner.
  • variants of an IL-1 cytokine family member can be made and evaluated using a display-based system or other library based screening.
  • the protein variants can be displayed or expressed and evaluated for ability to bind to a receptor for the IL-1 cytokine family member.
  • variants of IL-1 ⁇ , IL-1 Ra, or a cytokine domain described herein can be evaluated for ability to bind to the soluble extracellular domain of IL-1 Rl.
  • display-based systems include the following: for cell display, Chao et al. Nat Protoc. 2006;1 (2):755-68; Colby et al. Methods
  • IL-1 cytokine sequences as parental domains
  • other sequences can be used.
  • IL-1 cytokines e.g., from other species
  • Examples of such sequences from the UNIPROT database available at UniProt.org and see The UniProt Consortium, Nucleic Acids Res. D142-D148 (2010)), include the following:
  • Cytokine domains described herein can also include substitutions present in variant cytokine domains that are able to bind to IL-1 Rl.
  • position 15 of SEQ ID NO:1 (corresponding to position 20 of SEQ ID NO:3) can be Met or Asn.
  • Position 30 of SEQ ID NO:1 (corresponding to position 34 of SEQ ID NO:3) can be Gly, His, Trp, or Met.
  • variants of IL-1 ⁇ and IL-1 Ra include those described in Boraschi et al. (1996) Frontiers in Bioscience: A Journal and Virtual Library 1 , d270-308, Evans et al., J. Biol. Chem., 270:1 1477 (1995) and Greenfeder et al., J. Biol. Chem., 270:22460 (1995).
  • variants of IL-1 ⁇ include R1 1 G, R1 1 A, Q15H, E105G, and T147G. See, e.g., Evans et al.
  • Variants of IL-1 Ra include W16Y, Q20M, Q20N, Y34G, Y34H, Y34W, Y34M, Y147G, Y147H, Y147M, K145D, H54P, V18S, T108K, C1 16F, C122S, C122A, Y147G, H54P, H54I, and others in Evans et al. and Greenfeder et al., J. Biol. Chem., 270:22460 (1995).
  • a cytokine domain can include a residue identical to IL-1 Ra at one of the foregoing positions.
  • a cytokine domain can also include a residue differing from one of the foregoing mutations at a corresponding position.
  • IL-1 family cytokines can include one or more unpaired cysteine residues.
  • One or more, e.g., two, three or all such unpaired cysteine residues can be mutated to another amino acid, e.g., an uncharged amino acid such as alanine or serine.
  • P01 includes cysteines at positions 67, 70, 1 16, and 122 of SEQ ID NO:17.
  • One, two, three or all four such cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine.
  • P02 includes cysteines at positions 67, 70, 1 16, and 122 of SEQ ID NO:18.
  • cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine.
  • P03 includes cysteines at positions 70, 1 16, and 122 of SEQ ID NO:19.
  • One, two, or all three such cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine.
  • P04 includes cysteines at positions 70, 1 16, and 122 of SEQ ID NO:20.
  • One, two, or all three such cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine.
  • P05 includes cysteines at positions 8, 70, and 122 of SEQ ID NO:21 .
  • One, two, or all three such cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine.
  • An IL-1 family cytokine domain can also be cyclically permutated.
  • a C-terminal segment from the domain can be repositioned so that it is N-terminal to the original N-terminus and an N-terminal segment (generally comprising the remainder of the original protein after excision of the C-terminal segment).
  • the two repositioned segments can be separated by a linker (e.g., of between about three to ten amino acids). Generally, all the amino acids in the domain prior to permutation are retained except for the change in order.
  • the cut point for the permutation can be in a flexible region, e.g., a flexible loop such as such as the ⁇ 6- ⁇ 7 (e.g., amino acids corresponding to 71 -80 of SEQ ID NO:3) or the ⁇ 7- ⁇ 8 loop (e.g., amino acids corresponding to 84-99 of SEQ ID NO:3).
  • a flexible loop such as such as the ⁇ 6- ⁇ 7 (e.g., amino acids corresponding to 71 -80 of SEQ ID NO:3) or the ⁇ 7- ⁇ 8 loop (e.g., amino acids corresponding to 84-99 of SEQ ID NO:3).
  • a receptor binding agent described herein e.g., a receptor binding agent which includes an IL-1 family cytokine domain has a molecular weight less than 30, 25, 22, 20, 19, 18, or about 17 kDa. In some embodiments, the receptor binding agent has a molecular weight greater than 18, 19, 20, 22, 25, 30, 40, 45, or 50 kDa.
  • the receptor binding agent can include other polypeptide, polymeric or non-polymeric components, for example, components which modify the agents pharmacokinetics, stability, immunogenicity, and/or molecular weight.
  • the protein may include other modifications, e.g., post-translational or synthetic
  • the receptor binding agent is not glycosylated. In other embodiments, the receptor binding agent includes at least one glycosylation.
  • receptor binding agents described herein can include additional domains and features.
  • a receptor binding agent can be fused, directly or indirectly, to a domain of an antibody protein, e.g., to an Fc domain or to one or more constant domains (e.g., CH1 , CH2, or CH3).
  • the domains can be human domains or variants of human domains.
  • the Fc domain or the one or more constant domains can be located N-terminal or C-terminal to the receptor binding agent.
  • Fc domains can be obtained from any suitable immunoglobulin, e.g., from a human antibody, e.g., such as an antibody of the lgG1 , lgG2, lgG3, or lgG4 subtypes, IgA, IgE, IgD or IgM.
  • the Fc domain includes a sequence from an amino acid residue at about position Cys226, or from about position Pro230, to the carboxyl terminus of the Fc domain.
  • An Fc domain generally includes two constant domains, a CH2 domain and a CH3 domain, and optionally includes a CH4 domain.
  • a receptor binding agent includes a salvage receptor-binding epitope which increases in vivo serum half-life, as described, e.g., in US Patent
  • the epitope is include in an Fc region that is fused to the receptor binding agent.
  • a receptor binding agent includes a serum albumin sequence, or a portion of such sequence that binds to the FcRn receptor, or a sequence which binds to serum albumin, e.g., human serum albumin.
  • serum albumin e.g., human serum albumin.
  • certain peptides bind to serum albumin can be associated with the receptor binding agent, e.g., the sequence DICLPRWGCLW (SEQ ID NO:22). See also, Dennis et al. J. Biol. Chem. 277:35035-35043 (2002).
  • Receptor binding agents can be modified to include a sequence that increases the size and stability of the agent, e.g., a sequence described in WO2008/155134 or WO2009/023270. Such sequences can be generally biologically inactive, e.g., it does not modulate signaling mediated by IL-1 cytokine family members.
  • a variety of stabilizing polypeptide sequences can be used, e.g., sequences rich in glycine and/or serine, as well as other amino acids such as glutamate, aspartate, alanine or proline.
  • sequences can be designed to have at least 30, 40, 50, 60, 70, 80, 90 or 100% glycine and/or serine residues.
  • the combined length of stabilizing polypeptide sequences that are attached to a protein can be at least 20, 25, 35, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or more than 1000 or 2000 amino acids.
  • Stabilizing sequences can be, for example, fused to a biologically active polypeptide, for example to the N- or C-terminus of the receptor binding agent. Fusion of stabilizing sequences can result in a significant increase in the hydrodynamic radius of the fusion protein relative to the unmodified protein, which can be detected by ultracentrifugation, size exclusion chromatography, or light scattering, for example.
  • stabilizing sequences to contain few or none of the following amino acids: cysteine (to avoid disulfide formation and oxidation), methionine (to avoid oxidation), asparagine and glutamine (to avoid desamidation) and aspartate.
  • Stabilizing sequences can be designed to contain proline residues that tend to reduce sensitivity to proteolytic degradation. Binding Assays
  • the interaction of a receptor binding agent and its targets can be analyzed using any suitable approach, including for example radio-immunoassays, cell binding assays, and surface plasmon resonance (SPR).
  • SPR surface plasmon resonance
  • An exemplary cell binding assay using radio- iodinated protein competition is described in Boraschi, J. Immunol., 155(10):4719-25 (1995).
  • Binding affinities of a receptor binding agent for a cytokine receptor extracellular domain can be measured using SPR under approximately physiological conditions, e.g., 10 mM HEPES pH 7.4, 150 mM NaCI, 3 mM EDTA, 0.005% Tween-20.
  • Other methods that do not rely on SPR can also be used, e.g., to measure binding and affinity.
  • Information from binding assays can be used to provide an accurate and quanti- tative measure of the equilibrium dissociation constant (K D ), and kinetic parameters
  • K on and K off for the binding of a receptor binding agent to a target.
  • a receptor binding agent for the binding of a receptor binding agent to a target.
  • Such data can be used to compare different proteins, targets, and conditions.
  • This information can also be used to develop structure-activity relationships (SAR). For example, the kinetic and equilibrium binding parameters of variant proteins can be compared to the parameters of a reference or parent protein. Variant amino acids at given positions can be identified that correlate with particular binding parameters, e.g., high affinity and slow K off .
  • This information can be combined with structural modeling (e.g., using homology modeling, energy minimization, or structure determination by x-ray crystallography or NMR).
  • Proteins used for evaluating affinities can be produced in recombinant form and can include tags suitable for purification or immobilization, e.g., a FLAG tag, myc tag, hemagglutinin tag, His tag, or Fc domain fusion.
  • Extracellular domains of receptor proteins (such as IL-1 Rl, and IL-1 RAcP) can be produced in recombinant form by expression, e.g., in bacterial or insect cells, for example, using baculovirus expression in Sf9 cells.
  • Soluble receptor proteins can be immobilized in the BIAcore system, e.g., using chips that include reagents that bind to their tags, for example, a chip coated with IgG specific for the Fc domain or other tag.
  • receptor binding agents to function as receptor antagonists can be evaluated, e.g., in a cell based assay. For example, it is possible to evaluate IL-1 Rl inhibition by a receptor binding agent.
  • a cell based assay For example, it is possible to evaluate IL-1 Rl inhibition by a receptor binding agent.
  • assays for IL-1 activity are described in Boraschi et al. and include T cell proliferation assays, IL-6 and IL-8 production assays, and inhibition of calcium influx.
  • the ability of a receptor binding agent is evaluated for its ability to inhibit I L-1 ⁇ stimulated release of IL-6 from human fibroblasts.
  • Inhibition of IL- ⁇ ⁇ -stimulated cytokine release in MRC5 cells is correlated with the agent's ability to inhibit IL-1 mediated activity in vivo. Details of the assay are described in Dinarello et al., Current Protocols in Immunology, Ch. 6.2.1 -6.2.7, John Wiley and Sons Inc., 2000. Briefly, human MRC5 human fibroblasts (ATCC # CCL-171 , Manassas VA, USA) are grown to confluency in multi-well plates.
  • Cells are treated with titrated doses of the receptor binding agent and controls. Cells are subsequently contacted with 100 pg/ml of IL-1 ⁇ in the presence of the titrated agent and/or controls. Negative control cells are not stimulated with IL-1 ⁇ . The amounts of IL-6 released in each group of treated cells is measured using an IL-6 ELISA kit (e.g., BD Pharmingen, Franklin Lakes, NJ, USA). Controls that can be used include buffer alone, IL-1 Ra, and antibodies to IL-1 ⁇ .
  • mice are injected intraperitoneal ⁇ with titrated doses of the receptor binding agent and controls. Twenty-four hours after injection, mice are injected subcutaneously with recombinant human IL-1 ⁇ at a dose of 1 ⁇ g kg. Two hours after injection of the IL-1 ⁇ (peak IL-6 response time), mice are sacrificed, and blood is collected and processed for serum. Serum IL-6 levels are assayed by ELISA. Percent inhibition can be calculated based on the ratio of IL-6 detected in experimental animal serum to IL-6 detected in controls.
  • Receptor binding agents can be produced by expression in recombinant host cells, but also by other methods such as in vitro transcription and translation and chemical synthesis.
  • nucleic acids e.g., cDNA or genomic DNA
  • a replicable vector for cloning or for expression.
  • the vector may, for example, be a plasmid, cosmid, viral genome, phagemid, phage genome, or other autonomously replicating sequence.
  • the appropriate coding nucleic acid sequence may be inserted into the vector by a variety of procedures. For example, appropriate restriction endonuclease sites can be engineered (e.g., using PCR). Then restriction digestion and ligation can be used to insert the coding nucleic acid sequence at an appropriate location.
  • Vector components generally include one or more of an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence.
  • the receptor binding agent may be produced recombinantly either in isolation but also by fusion to one or more other components, such as a signal sequence, an epitope or purification moiety, and a label.
  • the receptor binding agent can include the pro domain of an interleukin-1 family member, e.g., which subsequently can be removed by proteolytic processing.
  • the receptor binding agent can be produced with or without a signal sequence.
  • it can be produced within cells so that it accumulates in inclusion bodies, or in the soluble fraction. It can also be secreted, e.g., by addition of a prokaryotic signal sequence, e.g., an appropriate leader sequence such as from alkaline phosphatase, penicillinase, or heat-stable enterotoxin II.
  • Exemplary bacterial host cells for expression include any transformable £ coli K-12 strain (such as £ coli BL21 , C600, ATCC 23724; £ coli HB101 NRRLB-1 1371 , ATCC-33694; £ coli MM294 ATCC-33625; £ coli W31 10 ATCC-27325), strains of B. subtilis, Pseudomonas, and other bacilli. Proteins produced in bacterial systems will typically lack glycosylation. Accordingly, in some embodiments, the receptor binding agents described herein are substantially free of glycosylation, e.g., free of glycosylation modifications of a mammalian or other eukaryotic cell.
  • the receptor binding agent can be expressed in a yeast host cell, e.g.,
  • the receptor binding agent can also be produced intracellular ⁇ or by secretion, e.g., using the yeast invertase leader or alpha factor leader (including Saccharomyces and Kluyveromyces forms), or the acid phosphatase leader, or the C. albicans glucoamylase leader (EP 362,179 published 4 Apr. 1990).
  • yeast invertase leader or alpha factor leader including Saccharomyces and Kluyveromyces forms
  • the acid phosphatase leader or the C. albicans glucoamylase leader
  • mammalian signal sequences may be used to direct secretion of the protein, such as signal sequences from secreted polypeptides of the same or related species, as well as viral secretory leaders.
  • the receptor binding agent can be produced with a pro domain of an interleukin-1 family member, e.g., an IL-1 a or IL-1 ⁇ pro domain.
  • Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Such sequences are well known for a variety of bacteria, yeast, and viruses.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria; the 2 ⁇ plasmid origin is suitable for yeast; and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells.
  • Selection genes typically contain a selection gene or marker.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies (such as the URA3 marker in Saccharomyces), or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • Various markers are also available for mammalian cells, e.g., DHFR or thymidine kinase.
  • DHFR can be used in conjunction with a cell line (such as a CHO cell line) deficient in DHFR activity, prepared and propagated as described by Urlaub et al., Proc. Natl. Acad. Sci. USA, 77:4216 (1980).
  • a cell line such as a CHO cell line
  • Expression and cloning vectors usually contain a promoter operably linked to the nucleic acid sequence encoding the receptor binding agent to direct mRNA synthesis.
  • promoters suitable for use with prokaryotic hosts include the ⁇ -lactamase and lactose promoter systems (Chang et al., Nature, 275:615 (1978); Goeddel et al., Nature, 281 :544 (1979)), alkaline phosphatase, a tryptophan (trp) promoter system (Goeddel, Nucleic Acids Res., 8:4057 (1980); EP 36,776), and hybrid promoters such as the tac promoter (deBoer et al., Proc. Natl.
  • Promoters for use in bacterial systems can also contain an appropriately located Shine-Dalgarno sequence.
  • the T7 polymerase system can also be used to drive expression of a nucleic acid coding sequence placed under control of the T7 promoter. See, e.g., the pET vectors (EMD Chemicals, Gibbstown NJ, USA) and host cells, e.g., as described in Novagen User Protocol TB053 available from EMD Chemicals and US 5,693,489.
  • such vectors can be used in combination with BL21 (DE3) cells and BL21 (DE3) pLysS cells to produce protein, e.g., at least 0.05, 0.1 , or 0.3 mg per ml of cell culture.
  • Other cells lines that can be used include DE3 lysogens of B834, BLR, HMS174, NovaBlue, including cells bearing a pLysS plasmid.
  • Exemplary promoters for use with yeast cells include the promoters for 3- phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem., 255:2073 (1980)) or other glycolytic enzymes (Hess et al., J. Adv. Enzyme Reg., 7:149 (1968); Holland,
  • yeast promoters are inducible and have the additional advantage of transcription controlled by growth conditions. Examples of inducible promoters include the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, metallothionein, and enzymes responsible for maltose and galactose utilization.
  • mRNA encoding a receptor binding agent from vectors in mammalian host cells can controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, and from heat-shock promoters.
  • viruses such as polyoma virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an
  • Heterologous promoter systems can also be used, e.g., promoters responsive to tetracycline. See Urlinger, S., et al. (2000) Proc. Natl. Acad. Sci. USA 97(14):7963- 7968. Transcription can also be driven by an enhancer sequence, located in cis or trans. Exemplary mammalian enhancer sequences include those for globin, elastase, albumin, oc-fetoprotein, and insulin.
  • Additional examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • the enhancer may be spliced into the vector at a position 5' or 3' to the coding sequence for the receptor binding agent, but is preferably located at a site 5' from the promoter.
  • Expression vectors used in eukaryotic host cells can also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5' and, occasionally 3', untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the receptor binding agent.
  • the expression vector may also include one or more intronic sequences.
  • the receptor binding agent can also be expressed in insect cells, e.g., Sf9 or SF21 cells, e.g., using the pFAST-BACTM system. Additional exemplary baculovirus expression vectors are available from Invitrogen, Life Technologies, Carlsbad CA, USA.
  • the receptor binding agent can also be expressed in mammalian cells. For example, cell lines of mammalian origin also may be employed.
  • mammalian host cell lines examples include the COS-7 line of monkey kidney cells (ATCC CRL 1651 ) (Gluzman et al., Cell 23:175, 1981 ), L cells, C127 cells, 3T3 cells (ATCC CCL 163), Chinese hamster ovary (CHO) cells, HeLa cells, and BHK (ATCC CRL 10) cell lines, and the CV1 /EBNA cell line derived from the African green monkey kidney cell line CV1 (ATCC CCL 70) as described by McMahan et al. (EMBO J. 10: 2821 , 1991 ).
  • Established methods for introducing DNA into mammalian cells have been described (Kaufman, R. J., Large Scale Mammalian Cell Culture, 1990, pp. 1569).
  • receptor binding agents can be recovered from culture medium, inclusion bodies, or cell lysates.
  • Cells can be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption, or cell lysing agents (e.g., detergents).
  • Receptor binding agents can be purified from other cell proteins or polypeptides that can be found in cell lysates or in the cell medium.
  • Various methods of protein purification may be employed and such methods are known in the art and described for example in Deutscher, Methods in Enzymology, 182 (1990); and Scopes, Protein Purification: Principles and Practice, Springer- Verlag, New York (2010) (ISBN:
  • Exemplary of purification procedures include: by fractionation on an ion- exchange column; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation-exchange resin such as DEAE; chromatofocusing; SDS-PAGE;
  • a purification method can include a combination of two different ion- exchange chromatography steps, e.g., cation exchange chromatograph followed by anion exchange chromatography, or vice versa.
  • Receptor binding agents can be eluted from ion exchange resin by a variety of methods include salt and/or pH gradients or steps.
  • the receptor binding agent includes a purification moiety (such as epitope tags and affinity handles). Such moieties can be used for affinity chromatography and can be optionally removed by proteolytic cleavage.
  • Ion exchange chromatography may also be used as an ion exchange separation technique. Ion exchange chromatography separates molecules based on differences between the overall charge of the molecules and can be used to separate intact form of receptor binding agents from other forms of such proteins.
  • Anionic or cationic substituents may be attached to matrices in order to form anionic or cationic supports for chromatography.
  • Anionic exchange substituents include diethylaminoethyl (DEAE), quaternary aminoethyl (QAE) and quaternary amine (Q) groups.
  • Cationic substitutents include carboxymethyl (CM), sulfoethyl (SE), sulfopropyl (SP), phosphate (P) and sulfonate (S).
  • Cellulose ion exchange resins such as DE23, DE32, DE52, CM-23, CM-32 and CM-52 are available from Whatman Ltd. (Maidstone, Kent, U.K).
  • SEPHADEXTM and other cross-linked ion exchangers are also known.
  • DEAE-, QAE-, CM-, and SP-SEPHADEXTM and DEAE-, Q-, CM- and S- SEPHAROSETM and SEPHAROSETM Fast Flow are available from Pharmacia AB.
  • DEAE and CM derivatized ethylene glycol-methacrylate copolymer such as
  • TOYOPEARL DEAE-650S or M and TOYOPEARL CM-650S or M are available from Toso Haas Co. (Philadelphia, PA, USA).
  • a cation exchange surface is an ion exchange surface with covalently bound negatively charged ligands, and which thus has free cations for exchange with cations in a solution in contact with the surface.
  • Exemplary surfaces include cation exchange resins, such as those wherein the covalently bound groups are carboxylate or sulfonate.
  • Commercially available cation exchange resins include CMC-cellulose, SP-SephadexTM and Fast S-SepharoseTM (Pharmacia).
  • An anion exchange surface is an ion exchange surface with covalently bound positively charged groups, such as quaternary amino groups.
  • An exemplary anion exchange surface is an anion exchange resin, such as DEAE cellulose, TMAE, QAE SephadexTM and Fast Q SepharoseTM (Pharmacia).
  • An exemplary purification scheme for a receptor binding agent includes lysing £. coli cells in lysis buffer following by depth filtration.
  • the material is then subject to cation exchange chromatography (CEX).
  • CEX eluate is then flowed over anion exchange media in an anion exchange chromatography (AEX) step.
  • AEX FT can be subject to a polishing step.
  • Material can then be processed by ultrafiltration/diafiltration, e.g., to concentrate or desalt the material.
  • Ultrafiltration/diafiltration membranes may be selected based on nominal molecular weight cut-off ("NMWCO") so as to retain the protein in the retentate, while allowing low molecular weight materials such as salts to pass into the filtrate.
  • Any buffering solution or sterile water may be used during the final buffer exchange step, e.g., depending on the desired final pH and conductivity of the product.
  • a receptor binding agent can be stored in a variety of solutions, including water,
  • Exemplary buffered solutions include sodium acetate pH 4.5, sodium acetate pH 4.7, sodium acetate pH 4.9, sodium acetate pH 5.1 , sodium acetate pH 5.3, sodium acetate pH 5.5, succinate pH 5.2, succinate pH 5.4, succinate pH 5.6, succinate pH 5.8, histidine pH 5.7, histidine pH 6.0, histidine pH 6.3, histidine 6.6, sodium phosphate pH 6.5, sodium phosphate pH 6.7, sodium phosphate 7.0, sodium phosphate pH 7.3, sodium phosphate pH 7.7, imidazole pH 6.5, imidazole pH 6.8, imidazole pH 7.2, Tris pH 7.0, Tris pH 7.5, Tris pH 7.7.
  • Buffering agents can be present, e.g., at a concentration of about 1 -100mM, 5-50mM, 10-50mM, or 5-25 mM.
  • the solution can further include a salt such as NaCI (e.g., 50 mM, 150 mM, or 250 mM, and ranges therebetween), arginine (e.g., at about 1%, 2%, 3%, 4%, 5%, 7.5%, and ranges therebetween), sucrose (e.g., at about 1 %, 2%, 3%, 4%, 5%, 7.5%, 8.5%, 10%, 15%, and ranges therebetween), and/or glycerol (e.g., at about 0.5%, 1 %, 2%, 3%, 4%, 5%, 7.5%, 8.5%, 10%, 15%, and ranges therebetween).
  • the receptor binding agent can be present in the composition in an amount of at least 50 mg, 100 mg, 500 mg, 1 g, 5 g, 10g, or
  • Host cell proteins refer to proteins in a preparation that differ from a receptor binding agent, and for example are endogenous proteins of the host cell from which the receptor binding agent was prepared, typically £ coli proteins. Preferably, host cell proteins are present at fewer than 10000, 1000, 900, 800, or 700 ppm (parts per million).
  • HCP can be detected, for example, by ELISA or other detection methods.
  • the ELISA can use polyclonal antibodies to HCPs.
  • An exemplary kit is available from Cygnus Technologies (CN# F410; Southport NC USA). Another exemplary kit uses AlphaScreen technology (AlphaLisa® E. coli HCP Kit, Product Number AL261 C/F, Perkin Elmer, Waltham MA USA).
  • Material containing or potentially containing a receptor binding agent can be evaluated, e.g., using high pressure liquid chromatography.
  • Exemplary analytical techniques include weak cation exchange high performance liquid chromatography (wCEX-HPLC), size exclusion high performance liquid chromatography (SE-HPLC), reverse phase liquid chromatography, ESI-MS, turbospray ionization mass spectrometry, nanospray ionization mass spectrometry, thermospray ionization mass spectrometry, sonic spray ionization mass spectrometry, SELDI-MS and MALDI-MS.
  • wCEX-HPLC weak cation exchange high performance liquid chromatography
  • SE-HPLC size exclusion high performance liquid chromatography
  • ESI-MS reverse phase liquid chromatography
  • turbospray ionization mass spectrometry nanospray ionization mass spectrometry
  • thermospray ionization mass spectrometry thermospray ionization mass spect
  • the N-terminal region of a receptor binding agent includes sequences identical to peptides from the N-terminal region of IL-1 ⁇ , for example, the peptide APVRS (SEQ ID NO:58).
  • Recombinant cells particularly £ coli cells
  • expressing such proteins can produce intact protein as well as other isoforms, including the des-Ala isoform and an isoform with an additional methionine (e.g., N- terminal to Ala1 ).
  • Receptor binding agents that include a proline at amino acid position 2 (where the N terminal residue is at position 1 ) can be susceptible to cleavage by £. coli proteases, such as aminopeptidase P which has cleavage specificity for X-PRO.
  • P03, P04, and P05 have a proline at position 2.
  • the intact forms of P03, P04, and P05 are 153 amino acids in length and begin with alanine whereas the des-Ala species are 152 amino acids in length and begin with proline.
  • Analytical techniques such as those above can be used to distinguish between intact forms and other forms, e.g., a des-Ala species.
  • An analytical exemplary technique is wCEX-HPLC.
  • P05 can be evaluated using a Dionex ProPac® WCX-10 4x250 mm column (Product Number 054993) as described in Example 9 below.
  • WCX- HPLC peaks can be evaluated by C4 reversed-phase (RP)-HPLC on-line with mass spectrometry. Intact P05 has theoretical mass: 1 7700.4 Da and is detectable as
  • the des-Ala species is detectable as 17629.4 Da, which is 71 Da less than the mass of the intact P05.
  • the 71 Da reduction in mass corresponds to removal of a single alanine residue.
  • a receptor binding agent can be formulated as a pharmaceutical composition.
  • the composition is sterile and includes one or more of a buffer, a
  • the composition can be an aqueous composition.
  • a receptor binding agent described herein can be formulated according to standard methods for a biologic. See e.g., Gennaro (ed.), Remington: The Science and Practice of Pharmacy, 20th ed., Lippincott, Williams & Wilkins (2000) (ISBN: 0683306472); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Ed.., Lippincott Williams & Wilkins Publishers (1999) (ISBN: 0683305727); Kibbe (ed.), Handbook of Pharmaceutical Excipients, 3rd ed. (2000) (ISBN: 091733096X); Protein formulation and delivery, McNally and Hastedt (eds.), Informa Health Care (ISBN: 0849379490) (2007).
  • a receptor binding agent for a pharmaceutical composition is typically at least 10,
  • the composition can contain the receptor binding agent at a concentration of between about 0.001 - 10%, e.g., 0.001 - 0.1 %, 0.01 - 1 %, or 0.1 % - 1 0%.
  • compositions can be substantially free of pyrogenic materials, substantially free of nucleic acids, and/or substantially free of cellular enzymes and components, such as polymerases, ribosomal proteins, and chaperone proteins.
  • a pharmaceutical composition can include a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • a “pharmaceutically acceptable salt” refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (see e.g., Berge, S.M., et al. (1977) J. Pharm. Sci. 66:1 -19), e.g., acid addition salts and base addition salts.
  • the receptor binding agent is formulated with one or more excipients, such as sodium chloride, and a phosphate buffer (e.g., sodium dibasic phosphate heptahydrate, sodium monobasic phosphate), and polysorbate. It can be provided, for example, in a buffered solution, e.g., at a concentration of about 5-100, 5- 30, 30-50, or 50-100 mg/ml and can be stored at 2-8 q C.
  • Pharmaceutical compositions may also be in a variety of other forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, and liposomes. The preferred form can depend on the intended mode of administration and therapeutic application. Compositions for the agents described herein are typically in the form of injectable or infusible solutions, or are for topical or ocular delivery (see below).
  • compositions typically are sterile and stable under the conditions of manufacture and storage. A pharmaceutical composition can also be tested to ensure it meets regulatory and industry standards for administration.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration.
  • Sterile injectable solutions can be prepared by incorporating an agent described herein in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating an agent described herein into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of an agent described herein plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • compositions can be engineered by inclusion of an agent that delays absorption, for example, monostearate salts and gelatin.
  • a receptor binding agent can be associated with a polymer, e.g., a substantially non-antigenic polymer, such as a polyalkylene oxide or a polyethylene oxide.
  • the polymer covalently attached to the receptor binding agent, e.g., directly or indirectly.
  • Suitable polymers will vary substantially by weight. Polymers having molecular number average weights ranging from about 200 to about 35,000 Daltons (or about 1 ,000 to about 15,000, and 2,000 to about 12,500) can be used.
  • a receptor binding agent can be conjugated to a water soluble polymer, e.g., a hydrophilic polyvinyl polymer, e.g. polyvinylalcohol or
  • polyvinylpyrrolidone A non-limiting list of such polymers include polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols,
  • polyoxyethylenated polyols such as polyoxyethylene, polyoxypropylene, and block copolymers of polyoxyethylene and polyoxypropylene (Pluronics);
  • polymethacrylates includee the saccharide monomers D-mannose, D- and L-galactose, fucose, fructose, D-xylose, L-arabinose, D-glucuronic acid, sialic acid, D-galacturonic acid, D-mannuronic acid (e.g.
  • polymannuronic acid or alginic acid
  • D-glucosamine D-galactosamine
  • D- glucose and neuraminic acid including homopolysaccharides and heteropolysaccharides such as lactose, amylopectin, starch, hydroxyethyl starch, amylose, dextrane sulfate, dextran, dextrins, glycogen, or the polysaccharide subunit of acid mucopolysaccharides, e.g. hyaluronic acid; polymers of sugar alcohols such as polysorbitol and polymannitol;
  • the receptor binding agent may be prepared with a carrier that will protect the compound against rapid release. It may be delivered as a controlled release formulation, delivered by an implant or a microencapsulated delivery system.
  • a carrier that will protect the compound against rapid release. It may be delivered as a controlled release formulation, delivered by an implant or a microencapsulated delivery system.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. See generally e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. Administration
  • a receptor binding agent can be administered to a subject, e.g., a human subject, by a variety of methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, intrasynovial, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural injection, intrasternal injection and infusion. Still other modes of administration include topical (e.g., dermal or mucosal) or inhalation (e.g., intranasal or intrapulmonary) routes. For many applications, the route of administration is one of: intravenous injection or infusion, subcutaneous injection, or intramuscular injection.
  • a receptor binding agent can be administered as a fixed dose or in a mg/kg dose. It can be administered intravenously (IV) or subcutaneously (SC).
  • the receptor binding agent can administered, for example, every day, every other day, every third, fourth or fifth day, every week, every three to five weeks, e.g., every fourth week, or monthly.
  • a pharmaceutical composition may include a "therapeutically effective amount" of an agent described herein.
  • a therapeutically effective amount of an agent may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual, e.g., amelioration of at least one disorder parameter, or amelioration of at least one symptom of the disorder (and optionally the effect of any additional agents being administered).
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the composition are outweighed by the therapeutically beneficial effects.
  • a receptor binding agent is typically administered in a therapeutically effective amount.
  • compositions can be administered using medical devices, e.g., implants, infusion pumps, hypodermic needles, and needleless hypodermic injection devices.
  • the device can include, e.g., one or more housings for storing pharmaceutical compositions, and can be configured to deliver unit doses of the receptor binding agent, and optionally a second agent.
  • the doses can be fixed doses, i.e., physically discrete units suited as unitary dosages for the subjects to be treated; each unit can contain a predetermined quantity of receptor binding agent calculated to produce the desired therapeutic effect in association with a pharmaceutical carrier and optionally in association with another agent.
  • a receptor binding agent can be administered to the subject having the disorder in an amount and for a time sufficient to induce a sustained improvement in at least one indicator that reflects the severity of the disorder.
  • An improvement is considered “sustained” if the subject exhibits the improvement on at least two occasions separated by one to four weeks.
  • the degree of improvement can be determined based on signs or symptoms, and can also employ questionnaires that are administered to the subject, such as quality-of-life
  • the baseline value for the chosen indicator or indicators is established by examination of the subject prior to administration of the first dose of the receptor binding agent. Preferably, the baseline examination is done within about 60 days of administering the first dose.
  • Improvement can be induced by repeatedly administering a dose of the receptor binding agent until the subject manifests an improvement over baseline for the chosen indicator or indicators.
  • the degree of improvement may be obtained by repeated administration over a period of at least a month or more, e.g., for one, two, or three months or longer, or indefinitely.
  • the agent can be administered for a period of one to six weeks or even a single dose.
  • treatment may be continued indefinitely at the same level or at a reduced dose or frequency. Treatment may also be discontinued, e.g., upon
  • a receptor binding agent such as one that binds to IL-1 Rl and that antagonizes IL-1 signaling, can be used to treat an "IL-1 mediated disorder," which includes any disease or medical condition that is (i) caused at least in part by IL-1 agonism, (ii) is associated with elevated levels or activity of an IL-1 signaling component (such as IL-1 a, IL-1 ⁇ , or IL-1 R I ) or elevated IL-1 signaling, and/or (iii) is ameliorated by decreasing IL-1 activity.
  • IL-1 mediated disorders include acute and chronic disorders, including autoimmune disorders and inflammatory disorders.
  • IL-1 mediated disorders include systemic and non-systemic disorders.
  • IL-1 a and IL-1 ⁇ are potent pro-inflammatory cytokines implicated in infectious responses as well as in inflammatory disease, including rheumatoid arthritis. Increased IL-1 production has been observed in patients with several autoimmune disorders, ischemia, and various cancers, therefore implicating IL-1 in these and related diseases.
  • treat refers to the administration of an agent described herein to a subject, e.g., a patient, in an amount, manner, and/or mode effective to improve a condition, symptom, or parameter associated with a disorder, e.g., a disorder described herein, or to prevent progression of a disorder, to either a statistically significant degree or to a degree detectable to one skilled in the art.
  • the treatment can be to intended to cure, heal, alleviate, relieve, alter, remedy, ameliorate, palliate, improve or affect the disorder, the symptoms of the disorder or the predisposition toward the disorder.
  • An effective amount, manner, or mode can vary depending on the subject and may be tailored to the subject. Exemplary subjects include humans, primates, and other non- human mammals.
  • a receptor binding agent can also be given prophylactically to reduce the risk of the occurrence of a disorder or symptom thereof.
  • the IL-1 mediated disorder can be an autoimmune disorder.
  • IL-1 mediated autoimmune disorders include: rheumatoid arthritis, ankylosing spondylitis, Behcet's syndrome, inflammatory bowel diseases (including Crohn's disease and ulcerative colitis), asthma, psoriasis, Type I diabetes, and other disorders identified herein.
  • a receptor binding agent described herein can be administered to a subject having or at risk for such IL-1 mediated autoimmune disorders.
  • the IL-1 mediated disorder can be an inflammatory disorder such as described below.
  • a receptor binding agent described herein can be administered to a subject having or at risk for such IL-1 mediated inflammatory disorders.
  • Exemplary IL-1 mediated disorders include:
  • Rheumatoid Arthritis and related arthritides can be used to treat a subject having or at risk for rheumatoid arthritis.
  • Rheumatoid arthritis is a chronic systemic autoimmune inflammatory disease that affects the synovial membrane in joints and which damages articular cartilage.
  • the pathogenesis of RA is T lymphocyte dependent and can include the production of autoantibodies known as rheumatoid factors.
  • Complexes of rheumatoid factor and antigen can form and accumulate in joint fluid and blood, inducing the infiltration of lymphocytes, neutrophils, and monocytes into the synovium.
  • Joints are typically affected in a symmetrical pattern, but extra-articular disease may also occur, e.g., causing pulmonary fibrosis, vasculitis, and cutaneous ulcers, or Felty's syndrome which manifests as neutropenia,
  • IL-1 Ra is indicated for moderately to severely active RA. See, e.g., Cohen, S. et al. Arthritis & Rheumatism 46, 614-24
  • Symptoms of active RA include fatigue, lack of appetite, low grade fever, muscle and joint aches, and stiffness. Muscle and joint stiffness are usually most notable in the morning and after periods of inactivity. During flares, joints frequently become red, swollen, painful, and tender, generally as a consequence of synovitis.
  • Scales useful for assessing RA and symptoms of RA include the Rheumatoid Arthritis Severity Scale (RASS; Bardwell et al., (2002) Rheumatology 41 (1 ):38-45), SF-36 Arthritis Specific Health Index (ASHI; Ware et al., (1999) Med. Care. 37(5 Suppl):MS40-50), Arthritis
  • a receptor binding agent described herein can be administered to a subject having or at risk for RA to delay onset and/or ameliorate one or more of the foregoing signs and symptoms.
  • the subject can have moderate to severe active RA.
  • the subject can be a non-responder to TNF inhibitor therapy (e.g., therapy with ENBREL®
  • the subject can have previously been administered a TNF inhibitor; or the subject can also be continuing to receive a TNF inhibitor (and responding or not responding).
  • the subject can also be administered methotrexate.
  • the subject can be administered one or more other DMARDS (disease modifying anti-rheumatic drugs), a corticosteroid, and/or a non-steroidal anti-inflammatory.
  • DMARDS disease modifying anti-rheumatic drugs
  • Still other drugs which can be co-administered with a receptor binding agent include inhibitors of CD28 (e.g., CTLA4- Ig), inhibitors of RANKL, IFNy, IL-6, IL-8, and IL-17.
  • Inhibitors include antibodies to such mediators, soluble receptors specific for such mediators, and/or antibodies to receptors for such mediators. Juvenile Chronic Arthritis.
  • a receptor binding agent can be used to treat juvenile chronic arthritis, e.g., in a subject that is less than 21 , 18, 17, 16, 15, or 14 years of age. Juvenile chronic arthritis resembles RA in several respects. Subjects can be rheumatoid factor positive. Subjects may have pauciarticular, polyarticular, or systemic forms of the disease. The arthritis can cause joint ankylosis and retarded growth and can also lead to chronic anterior uveitis and systemic amyloidosis. A receptor binding agent can be used delay onset of or ameliorate one or more such symptoms.
  • a receptor binding agent can be used to treat juvenile idiopathic arthritis, including systemic onset juvenile idiopathic arthritis (SO-JIA).
  • Subjects may have failed prior corticosteroid treatment or required corticosteroid treatment at a daily dose equal to or over 0.3 mg/kg. See e.g., Quartier et al. (201 1 ) Ann Rheum Dis. 70(5)747-54.
  • a receptor binding agent described herein can also be used to treat other rheumatic disorders including scleroderma, systemic lupus erythematosus, gout, osteoarthritis, polymyalgia rheumatica, psoriatic arthritis and chronic Lyme arthritis, inflammation of the voluntary muscle and other muscles, including dermatomyositis, inclusion body myositis, polymyositis, and lymphangioleimyomatosis, pyrogenic arthritis syndrome, pediatric granulomatous arthritis (PGA)/Blau syndrome, and other rheumatic disorders discussed herein.
  • PGA granulomatous arthritis
  • a receptor binding agent can be used to treat metabolic rheumatic disorders, e.g., disorders associated with hyperuricemia, e.g., gout including chronic acute gout, and other crystal-mediated arthropathies.
  • the agent can be used to treat drug-induced flares associated with gout, including for example flares induced by xanthine oxidase inhibitors, urate oxidase, or uricosuric agents.
  • crystals of uric acid can activate the inflammasome and trigger the release of I L- 1 ⁇ .
  • a receptor binding agent can be used to treat spondyloarthropathies, which include disorders such as ankylosing spondylitis, Reiter's syndrome, arthritis associated with inflammatory bowel disease, spondylitis associated with psoriasis, juvenile onset spondyloarthropathy and undifferentiated
  • Spondyloarthropathies are frequently associated with the HLA- B27 gene.
  • Subjects may lack rheumatoid factor and may exhibit sacroileitis with or without spondylitis and inflammatory asymmetric arthritis.
  • Subjects may also have ocular inflammation (see below).
  • a receptor binding agent can be used to treat scleroderma or systemic sclerosis.
  • Scleroderma is characterized by induration of the skin which may be localized or systemic.
  • Vascular lesions and endothelial cell injury in the microvascul- ature can also be present.
  • Subjects may exhibit mononuclear cell infiltrates in the cutaneous lesions and have anti-nuclear antibodies.
  • Other organs that show pathogenesis can include: the gastrointestinal tract which may have smooth muscle atrophy and fibrosis resulting in abnormal peristalsis/motility; the kidney which may have concentric subendothelial intimal proliferation affecting small arcuate and interlobular arteries with resultant reduced renal cortical blood flow, and which can cause
  • a receptor binding agent described herein can be administered to a subject having or at risk for scleroderma to ameliorate one or more of the foregoing signs and symptoms.
  • a receptor binding agent can be used to treat Sjogren's syndrome.
  • Sjogren's syndrome is characterized by immune-mediated inflammation and subsequent functional destruction of the tear glands and salivary glands.
  • the disease can be associated with or accompanied by inflammatory connective tissue diseases.
  • the disease is associated with autoantibody production against Ro and La antigens, both of which are small RNA-protein complexes. Lesions can result in
  • keratoconjunctivitis sicca keratoconjunctivitis sicca, xerostomia, with other manifestations or associations including biliary cirrhosis, peripheral or sensory neuropathy, and palpable purpura. Treatment of ocular disorders associated with Sjogren's is also discussed below.
  • Thyroid Disorders A receptor binding agent can be used to treat a thyroid disorder.
  • exemplary thyroid disorders include Graves' disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, and atrophic thyroiditis, and are the result of an autoimmune response against thyroid antigens with production of antibodies that react with proteins present in and often specific for the thyroid gland.
  • Experimental models are available including spontaneous models: rats (BUF and BB rats) and chickens (obese chicken strain) and inducible models created by immunization of animals with either thyroglobulin, thyroid microsomal antigen (thyroid peroxidase).
  • a receptor binding agent can be used to treat a diabetic disorder such as juvenile onset diabetes (including autoimmune diabetes mellitus and insulin-dependent types of diabetes) and maturity onset diabetes (including non-insulin dependent and obesity-mediated diabetes), type I diabetes and type II diabetes.
  • type I diabetes mellitus or insulin-dependent diabetes is associated with the autoimmune destruction of pancreatic islet cells caused by autoantibodies and auto-reactive T cells.
  • reducing IL-1 ⁇ activity can improve glucose control and beta-cell function and can be used to treat type II diabetes. See e.g. Owyang et al. Endocrinology. 2010;151 (6):2515-27.
  • a diabetic disorder such as juvenile onset diabetes (including autoimmune diabetes mellitus and insulin-dependent types of diabetes) and maturity onset diabetes (including non-insulin dependent and obesity-mediated diabetes), type I diabetes and type II diabetes.
  • type I diabetes mellitus or insulin-dependent diabetes is associated with the autoimmune destruction of pancreatic islet cells caused by autoanti
  • a receptor binding agent described herein can be administered to a subject that is not being administered insulin, e.g., the subject is not insulin dependent.
  • the subject can be pre-diabetic.
  • the subject can have either impaired glucose tolerance or impaired fasting glucose.
  • the subject can be obese or have a body mass index that is above 23, 25, 30, 35, or 40 kg/m 2 .
  • the subject can be insulin resistant, and/or characterized by hyperglycemia or hyperinsulinemia.
  • the subject can be at risk for progression to type II diabetes. See also Larsen, et al. (2007) NEJM 356:1517-26 and Larsen, et al. (2009). Diabetes Care 32:1663-8. In some aspects of the subject can be pre-diabetic.
  • the subject can have either impaired glucose tolerance or impaired fasting glucose.
  • the subject can be obese or have a body mass index that is above 23, 25, 30, 35, or 40 kg/m 2 .
  • the subject can be insulin resistant, and/or
  • the subject has a fasting plasma glucose of greater than 6.1 , 6.5, 7, or 8 mmol/L. In some embodiments, the subject has an A1 C level greater than 5.5, 5.7, 6, 6.4, 7, 7.5 or 8%.
  • the subject is also administered an insulin secretagogue, e.g., such as an sulfonylurea (e.g., chlorpropamide, tolazamide, acetohexamide, tolbutamide, glyburide, glimepiride, glipizide) and/or meglitinides (e.g., repaglinide, nateglinide) that stimulate insulin secretion.
  • an sulfonylurea e.g., chlorpropamide, tolazamide, acetohexamide, tolbutamide, glyburide, glimepiride, glipizide
  • meglitinides e.g., repaglinide, nateglinide
  • the receptor binding agent can be administered to reduce loss of and/or damage to pancreatic beta cells.
  • the subject is heterozygous or homozygous for the C allele of rs4251961 , located near the 5' of the gene IL1 RN.
  • Treatment with a receptor binding agent includes ameliorating or preventing deterioration of secondary conditions associated with diabetes, such as diabetic retinopathy, kidney transplant rejection in diabetic patients, obesity-mediated insulin resistance, and renal failure, which itself may be associated with proteinuria and hypertension.
  • diabetes such as diabetic retinopathy, kidney transplant rejection in diabetic patients, obesity-mediated insulin resistance, and renal failure, which itself may be associated with proteinuria and hypertension.
  • Receptor binding agents described herein can be used to treat a inflammatory gastrointestinal disorder, including for example coeliac disease, Crohn's disease, ulcerative colitis, idiopathic gastroparesis pancreatitis including chronic pancreatitis, acute pancreatitis, inflammatory bowel disease and ulcers including gastric and duodenal ulcers.
  • a receptor binding agent can be used to treat a pulmonary disease mediated by IL-1 .
  • Exemplary pulmonary diseases that can be treated include chronic obstructive pulmonary disease (e.g.
  • pulmonary alveolar proteinosis bleomycin-induced pneumopathy and fibrosis
  • pulmonary fibrosis including idiopathic pulmonary fibrosis and radiation-induced pulmonary fibrosis, pulmonary sarcoidosis, cystic fibrosis, collagen accumulation in the lungs, ARDS, broncho-pulmonary dysplasia (BPD), chronic obstructive pulmonary diseases, and chronic fibrotic lung disease of preterm infants.
  • the receptor binding agents described herein can be used to treat occupational lung diseases, including asbestosis, coal worker's pneumoconiosis, silicosis or similar conditions associated with long-term exposure to fine particles.
  • Inflammatory and fibrotic lung disease including eosinophilic pneumonia, idiopathic pulmonary fibrosis, and
  • hypersensitivity pneumonitis may involve a misregulated immune-inflammatory response that can be treated using a receptor binding agent.
  • Sarcoidosis is a condition of unknown etiology which is characterized by the presence of epithelioid granulomas in nearly any tissue in the body; involvement of the lung is most common.
  • the pathogenesis involves the persistence of activated macrophages and lymphoid cells at sites of the disease with subsequent chronic sequelae resultant from the release of locally and systemically active products released by these cell types.
  • a receptor binding agent described herein can be used to treat a cardiovascular disorder or injury, such as aortic aneurysms, acute coronary syndrome, arteritis, vascular occlusion, including cerebral artery occlusion, complications of coronary by-pass surgery, ischemia/reperfusion injury, heart disease, including atherosclerotic heart disease, myocarditis, including chronic autoimmune myocarditis and viral myocarditis, heart failure, including chronic heart failure, congestive heart failure, myocardial infarction, restenosis and/or atherosclerosis after heart surgery or after carotid artery balloon angioplastic procedures, silent myocardial ischemia, left ventricular pump dysfunction, post implantation complications of left ventricular assist devices, Raynaud's phenomena, thrombophlebitis, vasculitis including Kawasaki's vasculitis, veno-occlusive disease, giant cell arteritis, Wegener's granulomatosis, and Schoenle
  • Atherosclerosis or to reduce risk thereof.
  • IL-1 mediated signaling is activated by acute myocardial infarction and can initiate apoptotic cell death in the peri-infarct myocardial cells, extending the size of the infarct zone.
  • a receptor binding agent described herein can be administered to reduce damage caused by myocardial infarction.
  • the receptor binding agent can be administered a subject who is at risk for a myocardial infarction, or a subject who has experienced a myocardial infarction, particularly an acute myocardial infarction, e.g., within the last 2, 4, 6, 12, 24, or 48 hours.
  • the agent can be administered in
  • a receptor binding agent can also be used to treat stroke, sub-arachnoid hemorrhage, head trauma or brain injury, and/or inflammation associated with a cardiovascular disorder.
  • elevated levels of IL-1 ⁇ have been implicated neuroinflammation associated with stroke and brain injury (Rothwell, N. J., et al., TINS 23(12): 618-625, 2000).
  • the receptor binding agent can be administered to reduce such inflammation and other inflammation associated with ischemia and/or hypoxia.
  • the receptor binding agent can also be provided prophylactically, e.g., to reduce risk of such disorders and/or inflammation associated with such disorders.
  • the receptor binding agent can be administered a subject who is at risk for a stroke, an ischemic event, other cardiovascular event, or a hemorrhagic event (such as a subarachnoid hemorrhage), or a subject who has experienced a stroke, an ischemic event, other cardiovascular event, or a hemorrhagic event (such as a sub-arachnoid
  • hemorrhage e.g., within the last 2, 4, 6, 12, 24, or 48 hours.
  • Genitourinary and Renal Disorders disorders of the genitourinary system can also be treated with a receptor binding agent described herein. Such disorders include glomerulonephritis, including autoimmune glomerulonephritis, glomerulonephritis due to exposure to toxins or glomerulonephritis secondary to infections with haemolytic streptococci or other infectious agents.
  • Immune mediated renal diseases including glomerulonephritis and tubulointerstitial nephritis, are the result of antibody or T lymphocyte mediated injury to renal tissue either directly as a result of the production of autoreactive antibodies or T cells against renal antigens or indirectly as a result of the deposition of antibodies and/or immune complexes in the kidney that are reactive against other, non-renal antigens.
  • immune-mediated diseases that result in the formation of immune-complexes can also induce immune mediated renal disease as an indirect sequelae. Both direct and indirect immune mechanisms result in
  • a receptor binding agent can also be used to treat uremic syndrome and its clinical complications (for example, renal failure, anemia, and hypertrophic
  • cardiomyopathy including uremic syndrome associated with exposure to environmental toxins, drugs or other causes. Complications that arise from inflammation of the gallbladder wall that leads to alteration in absorptive function can also be treated.
  • cholelithiasis gallstones
  • choliedocholithiasis bile duct stones
  • a receptor binding agent can also be used to treat chronic pain conditions, such as chronic pelvic pain, including chronic prostatitis/pelvic pain syndrome, and post-herpetic pain.
  • a receptor binding agent described herein can be used to treat various forms of cancer, including acute myelogenous leukemia, chronic myelogenous leukemia, Epstein-Barr virus-positive nasopharyngeal carcinoma, glioma, colon, stomach, prostate, renal cell, cervical and ovarian cancers, lung cancer (SCLC and NSCLC), including cancer-associated cachexia, fatigue, asthenia, paraneoplastic syndrome of cachexia and hypercalcemia. See, e.g., Voronov et al. (2003) PNAS 100:2645-2650.
  • Solid tumors including sarcoma, osteosarcoma, and carcinoma, such as adenocarcinoma (for example, breast cancer) and squamous cell carcinoma are also treatable.
  • adenocarcinoma for example, breast cancer
  • squamous cell carcinoma are also treatable.
  • IL-1 ⁇ in certain tumors, see e.g., Krelin et al. (2007) Cancer Res. 67:1062-1071 .
  • Additional cancers include esophogeal cancer, gastric cancer, gall bladder carcinoma, leukemia, including acute myelogenous leukemia, chronic myelogenous leukemia, myeloid leukemia, chronic or acute
  • lymphoblastic leukemia and hairy cell leukemia can be treated with the receptor binding agents. See, e.g., Lust et al. (2009) Mayo Clin Proc 84(2):1 14-122.
  • a receptor binding agent can also be used to treat anemias and hematologic disorders, including chronic idiopathic neutropenia, anemia of chronic disease, aplastic anemia, including Fanconi's aplastic anemia; idiopathic thrombocytopenic purpura (ITP); thrombotic thrombocytopenic purpura, myelodysplastic syndromes (including refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, refractory anemia with excess blasts in transformation); myelofibrosis/myeloid metaplasia; and sickle cell vasocclusive crisis.
  • ITP idiopathic thrombocytopenic purpura
  • myelodysplastic syndromes including refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, refractory anemia with excess blasts in transformation
  • Autoimmune hemolytic anemia, immune pancytopenia, and paroxysmal nocturnal hemoglobinuria can result from production of antibodies that react with antigens expressed on the surface of red blood cells (and in some cases other blood cells including platelets as well) and is a consequence of the removal of those antibody coated cells via complement mediated lysis and/or ADCC/Fc-receptor-mediated mechanisms.
  • autoimmune thrombocytopenia including thrombocytopenic purpura
  • immune-mediated thrombocytopenia in other clinical settings, platelet
  • the receptor binding agent can also be administered to subjects having or at risk for various lymphoproliferative disorders, including autoimmune lymphoproliferative syndrome (ALPS), chronic lymphoblastic leukemia, hairy cell leukemia, chronic lymphatic leukemia, peripheral T-cell lymphoma, small lymphocytic lymphoma, mantle cell lymphoma, follicular lymphoma, Burkitt's lymphoma, Epstein-Barr virus-positive T cell lymphoma, histiocytic lymphoma, Hodgkin's disease, diffuse aggressive lymphoma, acute lymphatic leukemias, T gamma lymphoproliferative disease, cutaneous B cell lymphoma, cutaneous T cell lymphoma (i.e., mycosis fungoides) and Sezary syndrome.
  • ALPS autoimmune lymphoproliferative syndrome
  • chronic lymphoblastic leukemia hairy cell leukemia
  • chronic lymphatic leukemia chronic lymphatic leukemia
  • the receptor binding agents disclosed herein are also useful for treating conditions of the liver such as hepatitis, including acute alcoholic hepatitis, acute drug-induced or viral hepatitis, hepatitis A, B and C, sclerosing cholangitis, hepatic sinusoid epithelium, and inflammation of the liver due to unknown causes.
  • Receptor binding agents can also be used to treat disorders that involve hearing loss and that are associated with abnormal IL-1 expression.
  • disorders include cochlear nerve-associated hearing loss that is thought to result from an autoimmune process, e.g., autoimmune hearing loss, Meniere's syndrome and cholesteatoma, a middle ear disorder often associated with hearing loss.
  • Bone Disorders Non-arthritic disorders of the bones and joints and also treatable with the receptor binding agents described herein.
  • osteoclast disorders that lead to bone loss, such as but not limited to osteoporosis, including post-menopausal osteoporosis, osteoarthritis, periodontitis resulting in tooth loosening or loss, and prosthesis loosening after joint replacement (generally associated with an inflammatory response to wear debris), e.g., orthopedic implant osteolysis.
  • the receptor binding agents described herein can be used to treat primary amyloidosis and the secondary amyloidosis that is characteristic of various conditions, including Alzheimer's disease, secondary reactive amyloidosis; Down's syndrome; and dialysis-associated amyloidosis.
  • receptor binding agents can be used to treat Amyotrophic lateral sclerosis (ALS), Huntington's disease, and Parkinson's disease. These diseases can also involve formation of aggregates and amyloids that can trigger inflammatory responses.
  • Receptor binding agents can also be used to treat neuroinflammation and demyelinating diseases of the central and peripheral nervous systems, including multiple sclerosis; idiopathic demyelinating polyneuropathy or Guillain-Barre syndrome; and chronic inflammatory demyelinating polyneuropathy. These disorders are believed to have an autoimmune basis and result in nerve demyelination as a result of damage caused to oligodendrocytes or to myelin directly. In multiple sclerosis disease induction and involve T lymphocytes. Multiple sclerosis has either a relapsing-remitting course or a chronic progressive course.
  • T lymphocyte mediated, microglial cells and infiltrating macrophages are the predominant cell type at lesions.
  • the mechanism of oligodendrocyte cell death and subsequent demyelination is not known but is likely T lymphocyte driven.
  • Myopathies Receptor binding agents can be used to treat myopathies associated with inflammation and autoimmunity. Idiopathic inflammatory myopathies including dermatomyositis, polymyositis and others are disorders of chronic muscle inflammation of unknown etiology resulting in muscle weakness. Muscle
  • a receptor binding agent described herein can be used to treat a vasculitis disorder, e.g., a systemic vasculitis.
  • Systemic vasculitis includes diseases in which the primary lesion is inflammation and subsequent damage to blood vessels which results in ischemia/necrosis/degeneration to tissues supplied by the affected vessels and eventual end-organ dysfunction in some cases.
  • Vasculitides can also occur as a secondary lesion or sequelae to other immune-inflammatory mediated diseases such as rheumatoid arthritis, systemic sclerosis, etc., particularly in diseases also associated with the formation of immune complexes.
  • systemic necrotizing vasculitis polyarteritis nodosa, allergic angiitis and granulomatosis, polyangiitis;
  • Miscellaneous vasculitides include: mucocutaneous lymph node syndrome (MLNS or Kawasaki's disease), isolated CNS vasculitis, Behcet's disease, thromboangiitis obliterans (Buerger's disease) and cutaneous necrotizing venulitis.
  • MLNS mucocutaneous lymph node syndrome
  • CNS vasculitis isolated CNS vasculitis
  • Behcet's disease thromboangiitis obliterans
  • thromboangiitis obliterans Buerger's disease
  • cutaneous necrotizing venulitis The pathogenic mechanism of these vasculitis disorders is believed to be primarily due to the deposition of immunoglobulin complexes in the vessel wall and subsequent induction of an inflammatory response either via ADCC, complement activation, or both.
  • CAPS CAPS.
  • a receptor binding agent can be used to treat a CAPS disorder, i.e., CIAS1 Associated Periodic Syndromes.
  • CAPS includes three genetic syndromes:
  • Neonatal Onset Multisystem Inflammatory Disorder NOMID
  • Muckle-Wells Syndrome MFS
  • Familial Cold Autoinflammatory Syndrome FCAS
  • CAPS are inherited in an autosomal dominant manner with a sporadic or familial pattern.
  • CIAS1 encodes NALP3, a protein component of the "inflammasome", a subcellular enzyme complex that regulates the activity of caspase 1 .
  • IL-1 strongly induces the production of acute phase reactants in the liver, such as C-reactive protein (CRP) and serum amyloid A (SAA).
  • CRP C-reactive protein
  • SAA serum amyloid A
  • CAPS disorders share common clinical features and present as a spectrum of clinical severity. NOMID is the most seriously disabling, MWS somewhat less so and FCAS is the least severe. CAPS disorders have several overlapping features and individuals can have unique constellations of signs and symptoms. Features common to all these conditions include fevers, urticaria-like rash, arthritis or arthralgia, myalgia, malaise, and conjunctivitis.
  • MWS and NOMID are commonly associated with severe inflammation that may include the auditory system, meninges, and joints. These patients may suffer daily high spiking fevers and a chronic rash that frequently changes in distribution and intensity. Patients may suffer hearing loss or deafness. Conjunctivitis and papilledema are frequently observed. Amyloidosis may develop and lead to renal failure due to chronic inflammation and overproduction of acute phase reactants (particularly SM).
  • a receptor binding agent can be administered to a subject having NOMID, MWS, or FCAS or diagnosed as have a genotype associated with NOMID, MWS, or FCAS.
  • a receptor binding agent can be administered to a subject having TRAPS (TNF receptor associated periodic syndrome).
  • a receptor binding agent can be used to treat a dermatological disorder, such as an inflammatory dermatological disorder or an autoimmune or immune-mediated skin disease.
  • a dermatological disorder such as an inflammatory dermatological disorder or an autoimmune or immune-mediated skin disease.
  • An exemplary disorder is psoriasis. Additional autoimmune or immune-mediated skin disease including bullous skin diseases, erythema multiforme, and contact dermatitis are mediated by auto-antibodies, the genesis of which is T lymphocyte-dependent. Psoriasis is a T lymphocyte-mediated inflammatory disease. Lesions contain infiltrates of T lymphocytes, macrophages and antigen processing cells, and some neutrophils.
  • Additional disorders of the skin or mucous membranes that can be treated include acantholytic diseases, including Darier's disease, keratosis follicularis and pemphigus vulgaris. Further additional disorders include: acne, acne rosacea, alopecia areata, aphthous stomatitis, bullous pemphigoid, burns, eczema, erythema, including erythema multiforme and erythema multiforme bullosum (Stevens-Johnson syndrome), inflammatory skin disease, lichen planus, linear IgA bullous disease (chronic bullous dermatosis of childhood), loss of skin elasticity, mucosal surface ulcers, including gastric ulcers, neutrophilic dermatitis (Sweet's syndrome), dermatomyositis, pityriasis rubra pilaris, psoriasis, pyoderma gangrenosum, multicentric reti
  • a receptor binding agent can be used to treat an allergic disorder, such as asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity, allergic conjunctivitis (see also below) and urticaria. These diseases are frequently mediated by T lymphocyte induced inflammation, IgE mediated-inflammation, or both.
  • Asthma is a chronic condition involving the respiratory system in which the airway occasionally constricts, becomes inflamed, and is lined with excessive amounts of mucus, often in response to one or more triggers. Episodes may be triggered by such things as exposure to an environmental stimulant (or allergen) such as cold air, warm air, moist air, exercise or exertion, emotional stress, and viral illness. Airway narrowing causes symptoms such as wheezing, shortness of breath, chest tightness, and coughing.
  • a receptor binding agent can be used to treat asthma and can be formulated for topical or pulmonary delivery for such treatment or can be delivered parenterally.
  • a receptor binding agent can be administered to a subject that is about to undergo, is undergoing, or is recovering from a transplantation.
  • Transplantation associated diseases including graft rejection and graft-versus-host- disease (GVHD) are T lymphocyte-dependent; inhibition of T lymphocyte function is ameliorative.
  • Corneal transplantation can be associated with neovascularization which can be ameliorated by treatment with a receptor binding agent.
  • a receptor binding agent can be also used to treat complications resulting from solid organ transplantation, such as heart, liver, skin, kidney, lung (lung transplant airway obliteration) or other transplants, including bone marrow transplants.
  • the receptor binding agents described herein are useful for treating protozoal diseases, including malaria and schistosomiasis and to treat erythema nodosum leprosum; bacterial or viral meningitis; tuberculosis, including pulmonary tuberculosis; and pneumonitis secondary to a bacterial or viral infection including influenza infection and infectious mononucleosis.
  • a receptor binding agent is administered to a subject to reduce activity or expression of IL-6 in the subject.
  • the subject can have a disorder that is associated or mediated at least in part by IL-6.
  • the receptor binding agents described herein can be used to treat ocular disorders, including ocular disorders affecting the surface of the eye, inflammatory ocular disorders, and ocular disorders mediated at least in part by an autoimmune reaction.
  • the ocular disorder is a dry eye disorder that affects the surface of the eye.
  • the disorder includes conditions also referred to keratoconjunctivitis sicca, keratitis sicca, sicca syndrome, xerophthalmia, tear film disorder, decreased tear production, aqueous tear deficiency, and Meibomian gland dysfunction.
  • Dry eye can include forms that are associated with Sjogren's syndrome (SS), e.g., Sjogren's syndrome associated keratoconjunctivitis sicca, but also forms that are not so
  • SS Sjogren's syndrome
  • IL-1 has been implicated in the pathogenesis of dry eye disorders. See, e.g., Enriquez de Salamanca et al. (2010), Mol. Vis. 16:862-873.
  • Subjects having a dry eye syndrome can exhibit inflammation of the eye dry, and can experience scratchy, stingy, itchy, burning or pressured sensations, irritation, pain, and redness. Dry eye can be associated with both excessive eye watering and conversely insufficient tear production.
  • a receptor binding agent can be administered to such subjects to ameliorate or prevent the onset or worsening of one or more such symptoms.
  • a receptor binding agent can also be used to mitigate pain, e.g., ocular pain, such as due to neuroinflammation, in a subject who is experiencing such pain.
  • the ocular disorder is an ocular disorder associated with a systemic autoimmune disorder (such as Sjogren's syndrome and rheumatoid arthritis) or with a disorder associated with IL-1 or another IL-1 cytokine family member.
  • a systemic autoimmune disorder such as Sjogren's syndrome and rheumatoid arthritis
  • the patient may or may not have a systemic autoimmune disorder or other manifestations of a systemic autoimmune disorder.
  • a receptor binding agent can also be used to treat other disorders affecting the surface of the eye, such as the cornea. Such disorders include corneal ocular surface inflammatory conditions, corneal neovascularization, keratitis, including peripheral ulcerative keratitis and microbial keratitis.
  • the receptor binding agent can be used to treat a subject undergoing corneal wound healing (e.g., a subject having a corneal wound).
  • a receptor binding agent can be administered to a subject who is about to receive, undergoing, or recovering from a procedure involving the eye, e.g., corneal transplantation/ keratoplasty, keratoprosthesis surgery, lamellar transplantation, selective endothelial transplantation.
  • a receptor binding agent can be used to treat disorders affecting the conjunctiva, including conjunctival scarring disorders and conjunctivitis.
  • the receptor binding agent can be used to treat still other disorders such as pemphigoid syndrome and Stevens-Johnson syndrome.
  • a receptor binding agent described herein can be administered to a subject to modulate neovascularization in or around the eye. See, e.g., Dana (2007) Trans Am Ophthalmol Soc 105: 330-43.
  • a receptor binding agent can be administered to a subject having an allergic reaction affecting the eye, e.g., a subject experiencing severe allergic (atopic) eye disease such as allergic conjunctivitis.
  • the receptor binding agent can be administered topically. See also, e.g., Keane-Myers AM et al. (1999) Invest Ophthalmol Vis Sci, 40(12): 3041 -6.
  • a receptor binding agent can be administered to a subject having an autoimmune disorder affecting the eye.
  • autoimmune ocular disorders include sympathetic ophthalmia, Vogt-Koyanagi Harada (VKH) syndrome, birdshot retinochoriodopathy, ocular cicatricial pemphigoid, Fuchs' heterochronic iridocyclitis, and various forms of uveitis.
  • VKH Vogt-Koyanagi Harada
  • a receptor binding agent can be administered to a subject to treat any of the foregoing disorders.
  • a receptor binding agent can be administered to a subject who has or is at risk for diabetic retinopathy. See, e.g., Demircan et al. (2006) Eye 20:1366-1369 and Doganay et al. (2006) Eye, 16:163-170
  • Uveitis includes acute and chronic forms and includes inflammation of one or more of the iris, the ciliary body, and the choroid.
  • Chronic forms may be associated with systemic autoimmune disease, e.g., Behcet's syndrome, ankylosing spondylitis, juvenile rheumatoid arthritis, Reiter's syndrome, and inflammatory bowel disease.
  • systemic autoimmune disease e.g., Behcet's syndrome, ankylosing spondylitis, juvenile rheumatoid arthritis, Reiter's syndrome, and inflammatory bowel disease.
  • anterior uveitis inflammation is primarily in the iris (also ulceris).
  • Anterior uveitis can affect subjects who have systemic autoimmune disease, but also subjects who do not have systemic autoimmune disease.
  • Intermediate uveitis involves inflammation of the anterior vitreous, peripheral retina, and ciliary body, often with little anterior or chorioretinal inflammation.
  • Pan planitis results from inflammation
  • Posterior uveitis involves the uveal tract and primarily the choroid, and is also referred to as choroiditis.
  • Posterior uveitis can be associated with a systemic infection or an autoimmune disease. It can persist for months and even years.
  • a receptor binding agent can be administered to a subject to treat any of the foregoing forms of uveitis.
  • a receptor binding agent is used to treat a subject having or at risk for age-related macular degeneration (AMD).
  • the receptor binding agent can be applied topically to the eye, injected (e.g., intravitreally) or provided systemically.
  • a receptor binding agent described herein can be administered by any mode to treat an ocular disease.
  • the agent can be delivered by a parenteral mode.
  • the agent can be delivered directly to the eye or in the vicinity of the eye.
  • the protein can be administered topically or intraocularly, e.g., as described below.
  • Ophthalmic formulations containing a receptor binding agent can be delivered for topical administration, e.g., for administration as a liquid drop or an ointment, or for implantation, e.g., into an anterior chamber of the eye or the conjunctival sac.
  • Liquid drops can be delivered using an eye dropper.
  • the receptor binding agent can be present at 0.0001 - 0.1 %, 0.001 -5%, e.g., 0.005-0.5%, 0.05 - 0.5%, 0.01 -5%, 0.1 -2% or 1 %-5% concentration.
  • the ophthalmic formulation is applied directly to the eye including topical application to the eyelids or instillation into the space (cul-de-sac) between the eyeball and the eyelids.
  • the ophthalmic formulation can be designed to mix readily with the lacrimal fluids and spread over the surfaces of the cornea and conjunctiva. With the usual technique of
  • the major portion of the drug is deposited in the lower fornix.
  • Capillarity, diffusional forces, and the blinking reflex drive incorporation of the drug in the precorneal film from which it penetrates into and through the cornea.
  • Ophthalmic formulations can also include one or more other agents, e.g., an antiinflammatory steroid such as rimexolone, loteprednol, medrysone and hydrocortisone, or a non-steroidal anti-inflammatory.
  • an antiinflammatory steroid such as rimexolone, loteprednol, medrysone and hydrocortisone, or a non-steroidal anti-inflammatory.
  • the steroid can be present at a concentration of 0.001 to 1 %. In some embodiments, no steroid is present.
  • the receptor binding agent is the only active agent in the formulation.
  • the formulation can also include one or more of the following components:
  • Tonicity agents can be used to adjust the tonicity of the composition, e.g., to that of natural tears.
  • potassium chloride, sodium chloride, magnesium chloride, calcium chloride, dextrose and/or mannitol may be added to achieve an appropriate tonicity, e.g., physiological tonicity.
  • Tonicity agents can be added in an amount sufficient to provide an osmolality of about 150-450 mOsm or 250-350 mOsm.
  • the formulation can also include buffering suitable for ophthalmic delivery.
  • the buffer can include one or more buffering components (e.g., sodium phosphate, sodium acetate, sodium citrate, sodium borate or boric acid) to changes in pH especially under storage conditions.
  • the buffer can be selected to provide a target pH within the range of pH 6.0-7.5, e.g., 6.5-7.5.
  • the formulation can include an aqueous or phospholipid carrier.
  • the formulation can include agents to provide short-term relief, e.g., compounds which lubricate the eye and assist in tear formation.
  • phospholipid carriers which include one or more phospholipids
  • artificial tears compositions useful as artificial tears carriers include commercial products such as Tears NaturaleTM (Alcon Labs, Inc., TX USA).
  • the formulation can include: 1 mg dextran, 70 and 3 mg hydroxypropyl methylcellulose, and optionally a preservative such POLYQUAD®
  • phospholipid carrier formulations include those disclosed in US 4,804,539, US 4,883,658, US 5,075,104, US 5,278,151 , and US 5,578,586.
  • the formulation can also include other compounds that act as a lubricant or wetting agent.
  • viscosity agents such as: monomeric polyols, such as, glycerol, propylene glycol, ethylene glycol; polymeric polyols, such as polyethylene glycol, various polymers of the cellulose family: hydroxypropylmethyl cellulose ("HPMC"), carboxy methylcellulose sodium, hydroxy propylcellulose (“HPC”), dextrans, such as dextran 70; water soluble proteins, such as gelatin; and vinyl polymers, such as polyvinyl alcohol, polyvinylpyrrolidone, povidone and carbomers, such as carbomer 934P, carbomer 941 ; carbomer 940, carbomer 974P.
  • HPMC hydroxypropylmethyl cellulose
  • HPC hydroxypropylmethyl cellulose
  • HPC carboxy methylcellulose sodium, hydroxy propylcellulose
  • dextrans such as dextran 70
  • water soluble proteins such as ge
  • Still additional examples include polysaccharides, such as hyaluronic acid and its salts and chondroitin sulfate and its salts, and acrylic acid polymers.
  • the formulation has a viscosity between 1 to 400 cP.
  • the formulation can be packaged for single or multi-dose use, e.g., in a bottle with an associated dropper or as a set of single-use droppers.
  • the formulation can include one or more preservatives, e.g., to prevent microbial and fungal contamination during use.
  • exemplary preservatives include: benzalkonium chloride, chlorobutanol, benzododecinium bromide, methyl paraben, propyl paraben, phenylethyl alcohol, edetate disodium, sorbic acid, and polyquaternium-1 , and can be included at a concentration of from 0.001 to 1 .0% w/v. It is also possible to provide a formulation containing a receptor binding agent that is sterile yet free of preservatives. The formulation can be prepared for single use application.
  • Ophthalmic packs may be used to give prolonged contact of an ophthalmic formulation with the eye.
  • a cotton pledget is saturated with the formulation and then inserted into the superior or inferior fornix.
  • a receptor binding agent may also be administered by the way of iontophoresis. This procedure keeps the solution in contact with the cornea in an eyecup bearing an electrode. Diffusion of the drug is effected by difference of electrical potential.
  • a receptor binding agent may also be delivered by injection, e.g., subconjunctival injection.
  • the formulation can be injected underneath the conjunctiva facilitating passage through the sclera and into the eye by simple diffusion.
  • the formulation can also be injected underneath the conjunctiva and the underlying Tenon's capsule in the more posterior portion of the eye to deliver the agent to the ciliary body, choroid, and retina.
  • the formulation may also be administered by retrobulbar injection.
  • subjects can be evaluated using one or more of the following approaches: the Ocular Surface Disease Index (OSDI), corneal and conjunctival staining, and the Schirmer test.
  • OSDI Ocular Surface Disease Index
  • corneal and conjunctival staining corneal and conjunctival staining
  • Schirmer test the Ocular Surface Disease Index
  • the Ocular Surface Disease Index is a 12-item questionnaire that provides a rapid assessment of the symptoms of ocular irritation consistent with ocular surface inflammatory disorders, including DES, and their impact on vision-related functioning. See e.g. Ocul Immunol Inflamm. 2007 Sep-Oct;15(5):389-93.
  • the 12 items of the OSDI questionnaire are graded on a scale of 0 to 4. Scores are derived based on responses to provide an OSDI score on a scale of 0 to 100, with higher scores representing greater disability. A negative change from baseline indicates an
  • Corneal staining is a measure of epithelial disease, or break in the epithelial barrier of the ocular surface, typically seen with ocular surface inflammatory disorders such as dry eye. Corneal staining can exist even without clinically evident dry eye, if there is significant lid disease, such as posterior blepharitis. Corneal staining is highly correlated with ocular discomfort in many, though not all patients; in general corneal staining is associated with high scores in the OSDI, as described above.
  • saline-moistened fluorescein strips or 1 % sodium fluorescein solution are used to stain the tear film.
  • the entire cornea is then examined using slit-lamp evaluation with a yellow barrier filter (#12 Wratten) and cobalt blue illumination. Staining is graded according to the Oxford Schema. Conjunctival staining is likewise a measure of epithelial disease or break in the epithelial barrier of the ocular surface. Conjunctival staining is performed under the slit-lamp using lissamine green.
  • Schirmer Test The Schirmer test is performed in the presence and in the absence of anesthesia by placing a narrow filter-paper strip (5 x 3 5mm strip of
  • a receptor binding agent can be formulated for inhalatory or other mode of pulmonary delivery, e.g., to administer the agent to a tissue of the respiratory tract, e.g., the upper and lower respiratory tract.
  • the three common systems that can be used to deliver agents locally to the pulmonary air passages include dry powder inhalers (DPIs), metered dose inhalers (MDIs) and nebulizers.
  • DPIs dry powder inhalers
  • MDIs metered dose inhalers
  • nebulizers may be used to deliver receptor binding agents in a solubilized form or as a dispersion.
  • MDIs include a freon or other relatively high vapor pressure propellant that forces aerosolized medication into the respiratory tract upon activation of the device.
  • Nebulizers form a medicament aerosol to be inhaled by imparting energy to a liquid solution.
  • the agent can be stored in a lyophilized form (e.g., at room temperature) and reconstituted in solution prior to inhalation.
  • Direct pulmonary delivery of drugs during liquid ventilation or pulmonary lavage using a fluorochemical medium are also possible delivery modes. These and other methods can be used to deliver receptor binding agent.
  • the agent is delivered in a dosage unit form of at least about 0.02, 0.1 , 0.5, 1 , 1 .5, 2, 5, 10, 20, 40, or 50 mg/puff or more.
  • the receptor binding agent may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane,
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges for use in an inhaler or insufflator may be formulated containing a powder mix of the receptor binding agent and a suitable powder base such as lactose or starch, if the particle is a formulated particle.
  • other materials such as 100% DPPC or other surfactants can be mixed together to promote the delivery and dispersion of the formulated or unformulated receptor binding agent.
  • Particle size can also be varied to control whether deliver is to the lower or upper respiratory tract. For example, particles in the size range of 1 -5 microns or 10-50 microns can be used for the lower and upper respiratory tracts respectively.
  • a surfactant is generally a compound having a hydrophilic and lipophilic moiety, which promotes absorption of a drug by interacting with an interface between two immiscible phases.
  • Surfactants are useful in the dry particles for several reasons, e.g., reduction of particle agglomeration and reduction of macrophage phagocytosis.
  • Surfactants include phosphoglycerides, e.g., phosphatidylcholines, L-alpha-phosphatidylcholine dipalmitoyl (DPPC) and diphosphatidyl glycerol (DPPG); hexadecanol; fatty acids; polyethylene glycol (PEG); polyoxyethylene-9-; auryl ether; palmitic acid; oleic acid; sorbitan trioleate (Span 85); glycocholate; surfactin; poloxomer; sorbitan fatty acid ester; sorbitan trioleate; tyloxapol; and phospholipids.
  • phosphoglycerides e.g., phosphatidylcholines, L-alpha-phosphatidylcholine dipalmitoyl (DPPC) and diphosphatidyl glycerol (DPPG); hexadecanol; fatty acids; polyethylene glycol (PEG); polyoxyethylene-9-; auryl ether
  • antibodies that specifically recognize a chimeric cytokine domain described herein.
  • such antibodies preferentially bind to a chimeric domain relative to any parental cytokine domain.
  • a specific antibody can bind to an epitope that includes a junction between a segment from a first parental cytokine and a second parental cytokine.
  • a receptor-binding agent can be provided to a subject by delivering a nucleic acid that encodes and can express the receptor-binding agent.
  • a nucleic acid sequence encoding the receptor-binding agent can be placed under control of transcription control sequences and positioned in a nucleic acid vector for gene delivery, e.g., a viral vector.
  • exemplary viral vectors include adenoviral, retroviral, or adeno- associated viral vectors.
  • Vectors can be in the form of a plasmid or linear molecule, e.g., a linear double stranded DNA.
  • the delivered nucleic acid can be designed to be incorporated into the genome of the target cell, e.g., to integrate into the genome of the target cell. Alternatively, the delivered nucleic acid can be designed such that after delivery it exists autonomously in the cell.
  • Transcriptional control sequences can be engineered to provide transient or constitutive expression.
  • Transient control can include control regulated by an
  • exogenous agent e.g., by using transcriptional response elements for transcription factors responsive to exogenous agents (e.g., a steroid hormone or FK506) or environmental signals.
  • exogenous agents e.g., a steroid hormone or FK506
  • environmental signals e.g., environmental signals.
  • the delivered nucleic acid is generally engineered so that transcriptional and translational regulatory DNA are appropriately positioned relative to the coding sequence for the receptor-binding agent such that transcription is initiated and that protein is translated from the resulting message.
  • the nucleic acid can include transcriptional and translational regulatory nucleic acid sequences from mammalian cells, particularly humans. Such sequences include, e.g., promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, and enhancer or activator sequences.
  • the expression vector may include additional elements.
  • the expression vector can contain at least one or two sequences homologous to the host cell genome, e.g., flanking the expression construct.
  • the integrating vector may be directed to a specific locus in the host cell by selecting the appropriate homologous sequence for inclusion in the vector. Constructs for integrating vectors are well known in the art.
  • Exemplary adenoviral vectors include modified versions of human adenoviruses such as Ad2 or Ad5, in which the genetic elements necessary for the virus to replicate in vivo have been removed.
  • the E1 region can be removed and the genome can be further modified to accept an expression cassette coding for the receptor-binding agent.
  • retroviral vectors include LNL6, LXSN, LNCX, and lentiviral vectors.
  • lentiviral vectors are described by Pawliuk et al. (2001 ) Science 294:2368 and Imren et al. (2002) PNAS 99:14380 and include limited to, human immunodeficiency virus (e.g., HIV-1 , HIV-2), feline immunodeficiency virus (FIV), simian immunodeficiency virus (SIV), bovine immunodeficiency virus (BIV), and equine infectious anemia virus (EIAV).
  • human immunodeficiency virus e.g., HIV-1 , HIV-2
  • feline immunodeficiency virus FIV
  • SIV simian immunodeficiency virus
  • BIV bovine immunodeficiency virus
  • EIAV equine infectious anemia virus
  • These vectors can be constructed and engineered to be safe, e.g., by separating the essential genes (e.g., gag and pol) onto separate vectors and by rendering retrovirus replication defective.
  • the replication defective retrovirus is then packaged into virions through the use of a helper virus or a packaging cell line, by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel, F. M. et al. (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals.
  • the retroviral vector can include, in addition to sequences for expressing a receptor-binding agent, a left (5') retroviral LTR; a retroviral export element, optionally a reverse response element (RRE); a promoter, and a locus control region (LCR) or other transcriptional insulator sequence and a right (3') retroviral LTR.
  • Retroviral vectors can further contain a central polypurine tract (cPPT) or DNA flap to increase viral titers and transduction efficiency.
  • the nucleic acid containing a sequence encoding a receptor-binding agent can be delivered to any appropriate target cells, e.g., ex vivo or in vivo.
  • target cells include synovial cells, hematopoietic cells, dermal cells, and so forth.
  • the nucleic acid can be delivered to target cells associated with the eye, e.g., corneal epithelial cells. Delivery may include the debridement, or scraping of the corneal epithelium to expose a basal layer of epithelium. The nucleic acid for delivery is then added.
  • the nucleic acid is delivered to corneal endothelial cells, cells of the trabecular meshwork beneath the periphery of the cornea, cells of the choroid layer of the eye, cells of the retina, sclera or ciliary body, cells of the retinal or ocular vasculature, or cells of the vitreous body or cells of the lens, for example the lens epithelium.
  • Delivery methods include, e.g., retroviral infection, adenoviral infection, transformation with plasmids, transformation with liposomes containing exogenous nucleic acid, biolistic nucleic acid delivery (e.g., loading the nucleic acid onto gold or other metal particles and shooting or injecting into the cells), adeno-associated virus infection and Epstein-Barr virus infection. Delivery can be into cells or tissue by any method including needle injection, hypospray, electroporation, or a gene gun.
  • a receptor binding agent described herein can be administered with a second agent.
  • the two agents can be co-administered, or administered separately, e.g., using different regimes.
  • Exemplary second agents include an anti-inflammatory agent.
  • the second agent is an IL-17 antagonist (encompassing antagonists of all IL-17 family members, e.g., antagonists of IL-17A, IL-17F, IL-17B, IL-17C, IL-17D, and IL-17E).
  • IL-17 antagonists encompassing antagonists of all IL-17 family members, e.g., antagonists of IL-17A, IL-17F, IL-17B, IL-17C, IL-17D, and IL-17E).
  • Exemplary IL-17 antagonists include: agents (such as antibodies and other binding proteins) that bind to IL-17 (including IL-17A, IL-17F, IL-17B, IL-17C, IL-17D, and IL-17E) and which antagonize IL-17 mediated signaling; agents (such as antibodies and other binding proteins) that bind to one or more receptors for IL-17, such as IL-17RA and IL-17RC and which antagonize IL-17 mediated signaling; agents (such as antibodies and other binding proteins) that bind to a complex containing IL-17 and at least one receptor subunit, e.g., IL-17 and 11-17RA, or IL-17, IL-17RA, and IL-17RC and which antagonize IL-17 mediated signaling; and agents such as soluble receptors that include one or more of soluble extracellular domains of IL-17RA and IL-17RC and which antagonize IL-17 mediated signaling.
  • agents such as antibodies and other binding proteins that bind to
  • the second agent is an IL-12 antagonist.
  • IL- 12 antagonists include: agents (such as antibodies and other binding proteins) that bind to IL-12 (including p35 and p40) and which antagonize IL-12 mediated signaling; agents (such as antibodies and other binding proteins) that bind to one or more receptors for IL- 12, such as IL-12R 1 or IL-12R32 and which antagonize IL-12 mediated signaling; agents (such as antibodies and other binding proteins) that bind to a complex containing p35, p40 and at least one receptor subunit, e.g., IL-12R 1 or IL-12R32 and which antagonize IL-12 mediated signaling; and agents such as soluble receptors that include one or more of soluble extracellular domains of IL-12R31 or IL-12R32 and which antagonize IL-12 mediated signaling.
  • the second agent is an IL-23 antagonist.
  • exemplary IL- 23 antagonists include: agents (such as antibodies and other binding proteins) that bind to IL-23 (including p19 and p40) and which antagonize IL-23 mediated signaling; agents (such as antibodies and other binding proteins) that bind to one or more receptors for IL- 23, such as IL-12R 1 or IL-23R and which antagonize IL-23 mediated signaling; agents (such as antibodies and other binding proteins) that bind to a complex containing p19, p40 and at least one receptor subunit, e.g., IL-12R i or IL-23R and which antagonize IL- 23 mediated signaling; and agents such as soluble receptors that include one or more of soluble extracellular domains of IL-12R 1 or IL-23R and which antagonize IL-23 mediated signaling.
  • a receptor binding agent can be evaluated in an animal model for human disease, e.g., a human autoimmune and/or human inflammatory disease.
  • the agent can have specificity for the corresponding target protein in the animal.
  • a receptor binding agent can be evaluated in an animal model of rheumatoid arthritis, e.g., the collagen-induced arthritis (CIA) model.
  • CIA collagen-induced arthritis
  • Mclndoe et al. 1999, Proc. Natl. Acad. Sci. USA, 96:2210-2214; Issekutz, A. C. et al., Immunology (1996) 88:569; and Current Protocols in Immunology, Unit 15.5, Coligan et al. (eds.), John Wiley & Sons, Inc.
  • the model is produced by the immunization of susceptible strains of rat/mice with native type II collagen.
  • Collagen is emulsified in Complete Freund's Adjuvant (CFA) and injected intradermal ⁇ (100 ⁇ g collagen:100 ⁇ g CFA/mouse) at the base of the tail. Control mice are injected intradermal ⁇ with 0.05 ml of distilled water/CFA emulsion. A booster injection of collagen in incomplete adjuvant is given 21 days after the initial immunization. Disease is due to an autoimmune response induced upon immunization with collagen.
  • CFA Complete Freund's Adjuvant
  • the joints can be scored for arthritis, inflammation, pannus, cartilage damage and bone resorption using a defined scale.
  • the score for each limb is summed and recorded as the arthritic index (Al) for each individual animal.
  • Al arthritic index
  • EAE Experimental allergic encephalomyelitis
  • a receptor binding agent can be evaluated in the EAE model.
  • EAE is a T cell mediated autoimmune disorder characterized by T cell and mononuclear cell inflammation and subsequent demyelination of axons in the central nervous system. (See, for example, Bolton, C, 1995, Multiple Sclerosis, 143.)
  • a receptor binding agent can be evaluated in an animal model for skin allograft rejection, e.g., using murine tail-skin grafts.
  • Skin allograft rejection is mediated by T cells, helper T cells and killer-effector T cells.
  • T cells helper T cells
  • killer-effector T cells See, for example, Current Protocols in Immunology, Unit 4.4; Coligan et al. (eds.), 1995, John Wiley & Sons, Inc.
  • Other transplant rejection models can also be used. See, e.g., Tinubu et al., 1994, J.
  • An exemplary model for inflammatory bowel disease is the use of CD4+ CD45Rb-high cells transferred to SCID mice. See e.g., Hirano et al., J Pharmacol Sci. 2009 Jun;1 10(2):169-81 and the use of transgenic IL-10 deficient mice. See e.g., Inaba et al., Inflamm Bowel Dis., DOI: 10.1002/ibd.21253 (2010).
  • Yet another exemplary colitis model employs dextran sulfate sodium (DSS) to induce acute colitis.
  • colitis can be induced in mice by administration of 5% (wt/vol) DSS (molecular mass 30-40 kDa; ICN Biomedicals, Aurora, OH) in drinking water ad libitum. Symptoms resulting from this treatment bloody diarrhea, weight loss, colon shortening and mucosal ulceration with neutrophil infiltration. DSS-induced colitis is characterized histologically by infiltration of inflammatory cells into the lamina intestinal, with lymphoid hyperplasia, focal crypt damage, and epithelial ulceration. These changes are thought to develop due to a toxic effect of DSS on the epithelium and by phagocytosis of laminalitis cells and production of TNF-alpha and IFN-gamma. See, e.g., Hassan et al. PLoS One. 2010 Jan 25;5(1 ):e8868.
  • a receptor binding agent can be evaluated in a mouse model for dry eye disease.
  • Dry eye can be induced in mice by subcutaneous injection of scopolamine and then placement of the mice in controlled-environment chambers.
  • normal healthy 6 to 10 weeks old female C57BL/6 mice can be induced to have dry eye by continuous exposure to dry environment in a controlled environmental chamber.
  • the chamber has low relative humidity of less than 30% (generally about 19%), high airflow (15 liters/minute) and constant temperature (about 22 ⁇ C).
  • the mice placed in the chamber are also treated with scopolamine to inhibit tear secretion.
  • Sustained-release transdermal scopolamine patches can be obtained from Novartis (Summit, N.J.).
  • One-fourth of a patch is applied to the depilated mid-tail of mice every 48 hours.
  • the combination of the controlled environmental chamber and scopolamine produces severe dry eye in a relative short period of time (about 2-4 days).
  • the controlled environmental chamber can be prepared as described in Barbino et al., Invest. Ophthal. Vis. Sci., 46: 2766-271 1 (2005), and enables control of air flow, humidity, and temperature.
  • Mice can be monitored for signs of dry eye, e.g., by performing: a) cotton thread test to measure aqueous tear production, which is generally decreased in patients with dry eye; b) corneal fluorescein staining which is a marker of corneal surface damage; and general ophthalmic examination.
  • Cotton Thread Test Tear production can be measured with cotton thread test, impregnated with phenol red (Zone-Quick, Lacrimedics, Eastsound, Wash.). Under a magnifying fluorescent lamp, the thread is held with jeweler forceps and placed in the lateral cantus of the conjunctival fornix of the right eye for 30 or 60 seconds. The tear distance in mm is read under a microscope using the scale of a hemacytometer.
  • Corneal Fluorescein Staining can be evaluated by applying 1 .0 ⁇ of 5% fluorescein by a micropipette into the inferior conjunctival sac of the eye. The cornea is examined with a slit lamp biomicroscope using cobalt blue light 3 minutes after the fluorescein instillation. Punctuate staining is recorded in a masked fashion using a standardized National Eye Institute (NEI) grading system of 0-3 for each of the five areas in which the corneal surface has been divided.
  • NKI National Eye Institute
  • a receptor binding agent described herein can be used to detect IL-1 R1 in a sample, or cells expressing such a receptor.
  • the agent can be labeled directly or indirectly with a moiety that is a label or produces a signal, e.g., an enzyme, a radiolabel, an epitope, or a fluorescent protein (such as green fluorescent protein).
  • the agent can be contacted to a sample or to cells to determine if the receptor is present in the sample or on the cells, e.g., using standard immunoblotting, immunofluorescence, enzyme immunoassay (EIA), radioimmunoassay (RIA), fluorescence energy transfer, Western blot, and other diagnostic and detection techniques.
  • the receptor binding agent can also be labeled for in vivo detection and administered to a subject.
  • the subject can be imaged, e.g., by NMR or other tomographic means.
  • the binding agent can be labeled with a radiolabel such as 3 , ⁇ , 23 l, 99m Tc, 32 P, 25 l, 3 H, 4 C, and 88 Rh, fluorescent labels such as fluorescein and rhodamine, nuclear magnetic resonance active labels, positron emitting isotopes detectable by a positron emission tomography (“PET”) scanner,
  • PET positron emission tomography
  • chemiluminescers such as luciferin, and enzymatic markers such as peroxidase or phosphatase.
  • the agent can be labeled with a contrast agent such as paramagnetic agents and ferromagnetic or superparamagnetic (which primarily alter T2 response)
  • a receptor binding agent can also be used to purify cells which express the receptor to which it binds.
  • the receptor binding agent can be coupled to an immobilized support (e.g., magnetic beads or a column matrix) and contacted to cells which may express the receptor.
  • the support can be washed, e.g., with a physiological buffer, and the cells can be recovered from the support.
  • a receptor binding agent can also be used to purify soluble forms of the receptor to which it binds. For example, samples containing the soluble receptor can be contacted to immobilized receptor binding agent and then, e.g., after washing, can be recovered from the immobilized agent.
  • Nucleic acids encoding the proteins with the amino acid sequences listed in Table 4 were constructed in a pET vector containing a T7 promoter and ampicillin (pET31 series) or kanamycin resistance genes (pET28 series) (EMD
  • nucleic acid sequences encoding the above proteins are listed in Table 5.
  • the nucleic acid sequence further includes an ATG prior to the first nucleotide listed below.
  • the nucleic acid sequence further includes a stop codon (such as TAA, TAG, or TGA) after the last nucleotide listed below.
  • the proteins can include a range of different residues from IL-1 ⁇ and IL-1 Ra as illustrated below.
  • the cytokine domains can have 48-70% residues from I L- 1 ⁇ and 55-78% residues from IL-1 Ra. (Because a number of amino acid residues are conserved between the two proteins, the sum of the percentage identity to IL-1 ⁇ and to IL-1 Ra can be greater than 100%.) Table 6
  • Proteins that contain a hexa-histidine tag were expressed in E. coli cells
  • BL21 (DES) strain by induction with 1 mM IPTG at 37°C for 3 hours in LB broth media.
  • the cells were lysed in 20-50 mM Tris, 0.5 M NaCI, 2.5 mM EDTA, 0.1 % Triton X-100, pH 8.0. Lysates were subjected to IMAC chromatography using a HiTrap® pre-packed column (GE Healthcare, Piscataway NJ, USA).
  • the protein was loaded in 20 mM sodium phosphate, 0.5 M NaCI 10 mM imidazole, pH 7.4 buffer. It was eluted with 200 mM imidazole, 20 mM sodium phosphate, 0.5 M NaCI pH 7.4 buffer.
  • Eluted protein was dialyzed extensively against PBS, 0.1 % Polysorbate 80, pH 7.4, concentrated using an Amicon Ultra® (1 OK) filter, and stored at 4° or -80 °C. Proteins lacking a hexa-histidine tag were purified by ion exchange chromatography. P05 protein was purified by ion exchange chromatography. Lysate from expressing cells was applied to a GigaCapSTM column (Tosoh Bioscience LLC, King of Prussia, PA, USA) at low pH (approximately pH 5.5) in the absence of salt (conductivity approximately 1 mS/cm).
  • a 5 ml fraction containing the eluted protein was then diluted with 5 ml of H 2 0 and 5 ml of 20 mM Tris pH 8) and then applied to CaptoQTM resin (GE Healthcare, Piscataway NJ, USA) and eluted with a 0 mM to 250 mM NaCI gradient in 20 mM Tris pH 8.0.
  • the eluted protein was dialyzed extensively against 1 .25 X PBS 0.1 % TWEEN® 80 or 1 .25X PBS lacking TWEEN® and stored. See Fig. 6. P03 and P04 proteins were purified using similar methods.
  • Cells expressing P05 were also grown in TEKNOVATM Terrific Broth with animal free soytone (# T7660) supplemented with 10 g/L glucose, 10 mM MgS0 4 , trace elements (1 mg/ml TEKNOVATM 1000X Trace Elements, #T1001 ), and antibiotic in a Sartorius 2L BIOSTATTM A+ and were induced at OD 35-40 with 1 mM IPTG for about 6 hours.
  • Cells are grown at 37°C with 30% dissolved oxygen at pH 7.0, and agitation at 200-800 rpm with oxygen sparge at 2L/min. Cells are fed 9 g glucose/L/hr when glucose is depleted as detected by a pH increase. Feed is reduced to 6 g glucose/L/hr when the pH decreases (about 2.5 hrs after induction).
  • Cells were collected and lysed in lysis buffer (20 mM Tris, 10 mM EDTA, 0.1 % Triton, pH 8.0; 20 mM Tris, 10 mM EDTA, 0.1 % Triton, pH 7.0; 50 mM MOPS, 10 mM EDTA, 0.1 % Triton, pH 6.5; or 50 mM MOPS, 10 mM EDTA, 0.1 % Triton, pH 6.0).
  • lysis buffer (20 mM Tris, 10 mM EDTA, 0.1 % Triton, pH 8.0; 20 mM Tris, 10 mM EDTA, 0.1 % Triton, pH 7.0; 50 mM MOPS, 10 mM EDTA, 0.1 % Triton, pH 6.5; or 50 mM MOPS, 10 mM EDTA, 0.1 % Triton, pH 6.0).
  • Lysate is loaded onto Poros XS® cation ion exchange media (Life Technologies Corp., Carlsbad CA USA) at pH 5.3 and 3 mS/cm (35 mg product per ml column resin).
  • P05 protein is eluted by a step to pH 7.0 using buffer containing 100 mM MOPS 25 mM NaCI pH 7.0. The first eluting peak was discarded, and the second eluting peak was collected in pools and contained P05 protein. Early pools are enriched for intact P05 protein relative to a des-Ala species. This eluted material is then flowed over CaptoQTM anion exchange resin. The flow through, which contains intact P05 protein, is collected.
  • the media is washed with 100 mM MOPS 20 mM NaCI pH 6.0.
  • P05 protein is eluted by a step to pH 6.0 using buffer containing 100 mM MOPS 50-58 mM NaCI pH 6.0.
  • the first eluting peak was separated from subsequent peaks and contained intact P05 protein.
  • This eluted material is then flowed over CaptoQTM anion exchange resin. The flow through, which contains intact P05 protein, is collected.
  • the proteins or supernatants containing the proteins were evaluated in a cell- based assay for IL-1 activity.
  • HEK-BlueTM I L-1 ⁇ responsive cells were used to monitor IL-1 ⁇ activity (available from InvivoGen Inc., San Diego CA, USA). These cells include a SEAP reporter gene under the control of the IFN- ⁇ minimal promoter fused to five NF-KB and five AP-1 binding sites. IL-1 ⁇ engagement of IL-1 receptors on the cell surface lead to NF-KB activation and SEAP production.
  • the SEAP report can be detected, e.g., using QUANTI-BlueTM (InvivoGen Inc., San Diego CA, USA) and spectrophotometric analysis.
  • a HEK-Blue IL-1 ⁇ cell suspension was prepared from cells cultured to 70-80% confluence. The resuspended cells were adjusted to -330,000 cells/ml in fresh growth medium (DMEM, 4.5 g/l glucose, 2 mM L-Glutamine, 10% (v/v) heat-inactivated fetal bovine serum (30 min at 56°C), 50 U/ml penicillin, 50 ⁇ g/ml streptomycin, 100 ⁇ g/ml NormocinT).
  • DMEM fresh growth medium
  • penicillin 50 ⁇ g/ml streptomycin
  • 100 ⁇ g/ml NormocinT 100 ⁇ g/ml NormocinT.
  • Reagents were added to wells of a flat-bottom 96-well cell culture plate: 10 ⁇ of IL-1 ⁇ at 20 ng/ml, 1 ⁇ of the agent of interest and 30 ⁇ of cell culture medium to a final volume of 50 ⁇ . Positive and negative controls samples were prepared in parallel.
  • HEK-Blue IL-1 ⁇ cell suspension (-50,000 cells) was added to each well and the plate was cultured overnight at 37°C in 5% C0 2 tissue culture incubator.
  • the final IL-1 ⁇ concentration (in the 200 ⁇ final volume) was 0.1 ng/ml. I L-1 ⁇ activity was evaluated the next day (12-15 hours later). Prior to quantitation, the
  • QUANTI-BlueTM reagent was prepared according to the manufacturer's instructions.
  • a flat bottomed 96-well assay plate was prepared in which 150 ⁇ of QUANTI-BlueTM solution was added to each well.
  • 50 ⁇ of conditioned media from the wells of the 96 well tissue culture plate was added to each well of the assay plate.
  • the plate was incubated at 37°C for approximately 15-20 minutes. SEAP levels were then measured using a spectrophotometer at 620-655 nm.
  • FIG. 7B shows antagonism of IL-1 ⁇ activity by P01 at a range of IL-1 ⁇ protein concentrations using the HEKBlueTM cell assay above. Antagonism increased with increasing amounts of P01 (x-axis reflects microliters of supernatant containing P01 ).
  • the proteins P01 , P02, P03, P04, and P05 each antagonized IL-1 ⁇ activity. See FIG. 8A and 8B, for example.
  • the IC50 of P05 was less than about 5 ng/ml.
  • P05 was test for ability to agonize IL-1 Rl in this assay and was not observed to have any detectable agonistic activity even at the highest concentrations tested, 1 mg/ml.
  • P01 , P02, P03, P04, and P05 also inhibited IL-1 ⁇ induced IL-6 expression in MG-63 cells, a human osteosarcoma cell line that is responsive to IL-1 ⁇ .
  • hexa-histidine tagged P05 was observed to have biological activity. See also Example 8 below regarding untagged P05.
  • binding properties of proteins for soluble recombinant human IL-1 Rl were evaluated using surface plasmon resonance with a Reichert SR7000DC Dual Channel SPR system. Binding was evaluated in phosphate buffered saline with 0.005% Tween 20.
  • IL-1 ⁇ was observed to have a K D of between 8-9 nM and a dissociation constant (K d ) of between 2-3 ⁇ 1 0 "3 s ⁇ ⁇ and in another experiment a K D of about 2 nM, an association constant of 1 .3-1 .5 ⁇ 10 6 M ⁇ V, and a dissociation constant (K d ) of about 2.9-3.0 ⁇ 10 ⁇ 3 s "1 . See Fig. 9A.
  • the P01 protein bound with similar association kinetics as I L-1 ⁇ , but did not dissociate during of the dissociation phase of the binding experiment (about 1 80 seconds).
  • the P01 protein bound to IL-1 Rl with a greater affinity than did IL-1 ⁇ under similar conditions.
  • Binding of IL-1 Ra was observed to have a K D of about 0.33 nM, an association constant (K a ) of about 2 ⁇ 1 0 5 M ⁇ s ⁇ ⁇ and a dissociation constant (K d ) of about 6.6 ⁇ 1 0 ⁇ 5 s ⁇ . See Fig. 9B.
  • Chimeric cytokine domains P01 , P02, P03, P04, and P05 were observed to have K D ranging from about 1 2-1700 pM, an association constant (K a ) ranging from about 3 ⁇ 10 4 M ⁇ V to 3 ⁇ 10 6 M ⁇ s ⁇ ⁇ and a dissociation constant (K d ) ranging from about 2 ⁇ 10 ⁇ 5 to 1 ⁇ 10 ⁇ 3 s ⁇ . See for example Fig. 9C and 9D and Table 7 below. Table 7
  • Additional exemplary chimeric IL-1 family proteins also include the following:
  • the polypeptide below is a chimeric domain that includes at least two segments from IL-1 a and at least two segments from IL-1 Ra.
  • a circularly permuted IL-1 chimeric domain can be constructed by linking the N- terminus to the C-terminus of the molecule using a linker sequence and selecting a new location for each of the termini.
  • the linker length can be between five to ten, e.g., about seven amino acids.
  • Preferred locations for new termini are in loops which face away from the receptors, such as the ⁇ 6- ⁇ 7 loop (e.g., amino acids corresponding to 71 -80 of SEQ ID NO:1 ) or the ⁇ 7- ⁇ 8 loop (e.g., amino acids corresponding to 84-99 of SEQ ID NO:1 ).
  • circularly permuted IL-1 chimeric domains include: DKPTLQLESVDPKNYPKKKMEKRFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDE GVMVTKFYMQFVSSGGSGGGSAPVRSLNCRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMS FVQGEESNDKIPVALGLKEKNLYLSCVLKD (SEQ ID NO: 40) and
  • Proteins P03, P04, P05, mll_-1 Ra (methionyl IL-1 Ra), and IL-1 ⁇ were prepared in phosphate-buffered saline (PBS), pH 7.4, at 0.5 mg/ml.
  • the proteins were combined with SYPRO orange dye (Invitrogen, CA) at a 1 :500 dilution of the stock concentration and subject to differential scanning fluorimetry. See, e.g., He et al. (2010) J. Pharm. Sciences, 99 1707-1720. Fluorescence measurements were monitored using an Agilent Mx3005 QPCR machine as the temperature was increased from 25°C to 95°C at a rate of 1 °C per minute.
  • T m Melting temperature
  • P05 65 59 P04 has a T m that is about 4 degrees higher than mlL-1 Ra and IL-1 ⁇ and exhibits an onset of unfolding about 3 degrees higher than mlL-1 Ra and about 10 degrees higher than IL-1 ⁇ .
  • P03 and P05 have a T m that is about 9 degrees higher than mlL-1 Ra and IL-1 ⁇ and exhibit an onset of unfolding about 1 1 degrees higher than mlL-1 Ra and about 18 degrees higher than IL-1 ⁇ .
  • Purified P05 (lacking a hexa-histidine tag) was prepared in 1 .25x PBS and tested in a murine model of dry eye disease.
  • female C57BL/6 mice 6 to 10 weeks of age from Jackson Laboratories (acclimated 1 to 2 weeks in an animal holding room with ⁇ 30% relative humidity, hydrogel food supplement, and envirodry environment enrichment) were pre-screened for fluorescein staining on Day 0.
  • fluorescein staining freshly made fluorescein diluted in WFI H 2 0 at 10 mg/mL was administered at 0.4 ⁇ - to each eye. Approximately 8-13 minutes after administration, eyes were scored using an Olympus fluorescent dissecting microscope.
  • Punctuate staining was recorded using the standardized National Eye Institute (NEI) grading system of 0-3 for each of the five areas into which the corneal surface has been divided (score range 0-15/eye). Using a teaching bridge, two masked scorers evaluated mice at the same time to give a single collective score for each eye.
  • NKI National Eye Institute
  • mice with scores ⁇ 7 for each eye were placed in a dry eye chamber (20% ⁇ 2% humidity and constant air flow -21 L/min/cage) on day 1 and were maintained in this chamber during the course of the experiment (except for examination).
  • mice were scored again and randomized into treatment groups with 8 to 10 mice/group. Mice were randomized such that each cage of 4 to 5 mice had approximately the same mean disease score. Beginning on day 3 and after
  • mice were topically administered P05 or vehicle (1 .25X PBS) in an eye drop at 3 ⁇ /eye BID frequency. Mice were examined and scored on days 7, 9, and 1 1 for corneal fluorescein staining as described above. Scorers were blinded as to the treatment groups during the course of the experiment.
  • FIG. 1 1 A is a bar graph of mean corneal staining score ⁇ SEM at day 0, 3, 7, 9, and 1 1 for mice from two identical experiments under the following bid treatments: no treatment, vehicle (1 .25X PBS), and 10 mg/ml (1 %) P05. 10 mg/ml P05 significantly reduced corneal staining on days 7, 9, and 1 1 of the experiment. Efficacy as evaluated by a reduction in corneal staining was also observed with doses as low as 0.1 mg/ml P05. Recombinant IL-1 Ra produced in E. coli also moderately reduced corneal staining in the animal model.
  • the effect of 10 mg/ml P05 was specific based on a comparison to 10 mg/ml murine serum albumin in the same vehicle. No effect was seen with 10 mg/ml murine serum albumin (MSA) relative to vehicle, and the effect of 10 mg/ml P05 was statistically significant relative to 10 mg/ml murine serum albumin.
  • 10 mg/ml P05 was also compared to 0.05% cyclosporine in an ophthalmic emulsion (Restasis®). Whereas P05 reduced corneal staining, no effect was observed for the 0.05% cyclosporine ophthalmic emulsion after ⁇ 1 week of bid dosing.
  • a capture step has been developed for the purification of P05 produced by fermentation in BLR(DE3) E. coli cells.
  • the elution conditions were defined through a statistical design of experiment approach (DOE) on a 1 ml_ column.
  • DOE statistical design of experiment approach
  • the optimized conditions were performed on an intermediate scale (10 ml_ column).
  • the product is extracted by microfluidization in a lysis buffer consisting of 20 mM Tris at pH 7.0 with 10 mM EDTA added.
  • the clarified lysate is adjusted to a pH of 5.3 and a conductivity of 3 mS/cm.
  • the conditioned lysate is loaded onto a PorosXS (strong cation exchange resin) with a 2 min residence time to a capacity of 25-30 mg P05/ml_ column.
  • PorosXS strong cation exchange resin
  • the column is washed with equilibration buffer to remove unbound species.
  • a second wash at pH 6.0 and 3 mS/cm is implemented to remove a population of impurities.
  • the product is eluted at pH 6.0 and 6.6 mS/cm.
  • a product related species is eluted at pH 6.0 and 12.4 mS/cm.
  • the column is cleaned with a high salt buffer and NaOH.
  • P05 is a 17 kDa protein with a pi of 6.58.
  • the pi of the des-ALA species is 6.8 and is readily resolved by analytical weak cation exchange chromatography. This species is likely to be a product of aminopeptidase P activity found in E. coli.
  • the species can be identified by mass spectroscopy, peptide mapping, and HPLC methods.
  • PorosXS is based on a crosslinked poly(styrene- divinylbenzene) bead with a nominal 50 ⁇ particle size.
  • the chromatographic matrix has sulfopropyl surface chemistry and was designed to tolerate elevated levels of salt during binding.
  • the PorosXS (CN 4404339, Life Technologies) material was procured as a bulk slurry and packed into a 5x50 mm Tricorn column (CN 28-4064-09, GE Healthcare) with a final column volume of 1 ml_ (5 cm bed height) or into a 1 0x 1 50 mm Tricorn column (CN 28-4064-16, GE Healthcare) packed to a final volume of 10.5 ml_ (13.4 cm bed height).
  • Buffers All buffers were prepared with MilliQ water volumetrically. To reach the desired pH, either 10 N HCI or 1 M NaOH (made from a stock solution of 10 M NaOH) was added to titrate the buffer. After preparation, all buffers were filtered through 0.2 ⁇ PES bottle top filters. All pH and conductivity measurements were performed at room temperature ( ⁇ 20-25°C).
  • PorosXS Buffers CEX equilibration buffer - 10 mM acetic acid (HoAC) with 21 mM NaCI made by adding 0.57 ml_ of glacial acetic acid and 2.44 g of NaCI per L of buffer. The final pH was 5.3 and the final conductivity was 3 mS/cm.
  • HoAC acetic acid
  • MOPS free acid 1 .5 g of MOPS sodium salt, and 1 .28 g of NaCI per L of buffer.
  • the final pH was 5.7-6.6 (as desired) and the final conductivity was 3 mS/cm.
  • CEX elution buffer - 100 mM MOPS with 1 18 mM NaCI made by adding 19.5 g of MOPS free acid, 1 .5 g of MOPS sodium salt, and 6.93 g of NaCI per L of buffer.
  • the final pH was 5.7-6.6 (as desired) and the final conductivity was 12.4 mS/cm.
  • CEX strip buffer - 10 mM acetic acid with 3 M NaCI made by adding 0.57 ml_ of glacial acetic acid and 175 g of NaCI per L of buffer. The final pH was 5.3 and the final conductivity was 188 mS/cm.
  • Lysate preparation The pH of the lysate was adjusted using 200 mM acetic acid at pH 4.5 made by mixing 1 1 .5 ml_ of glacial acetic acid per L of buffer. The solution was titrated to a final pH of 4.5 using 1 M NaOH. This solution was used in order to avoid localized precipitation due to low pH of concentrated or strong acids.
  • the load material for these experiments was an extract from a 2 L fed-batch bioreactor run.
  • the extraction of the product from the cell pellet was performed using 20 mM Tris at pH 7.0 with 10 mM EDTA added.
  • the fresh extract was diluted to a conductivity of 3 mS/cm with MilliQ water (typically 1 :1 -1 .5 dilution).
  • the diluted extract was frozen at -20°C in 41 ml_ aliquots.
  • To prepare the load an aliquot was thawed at room temperature and titrated to pH 5.3 using 200 mM acetic acid at pH 4.5. Small adjustments in conductivity were made by addition of MilliQ water when needed after titration.
  • the load was diluted 2.5 ⁇ to a concentration of ⁇ 1 .7 g/L using CEX equilibration buffer.
  • the load was sterile filtered through a 0.8/0.2 ⁇ filter.
  • the conditioned load was used the same day as prepared.
  • HCP host cell protein
  • ELISA enzyme-linked immunosorbent assay
  • polyacrylamide gel electrophoresis SDS-PAGE
  • SDS-PAGE polyacrylamide gel electrophoresis
  • the running buffer is 1 ⁇ MES SDS running buffer prepared from a 20 ⁇ concentration (CN NP0002, Invitrogen).
  • Novex sharp prestained protein standards CN 57318, Invitrogen are used as molecular weight indicators.
  • Samples for analysis were prepared by dilution with MilliQ water to a final volume of 30 ⁇ _ and 10 ⁇ _ of 4x Lithium dodecyl sulfate (LDS) (CN NP0008, Invitrogen) was added to a final concentration of 1 ⁇ .
  • LDS Lithium dodecyl sulfate
  • the samples were mixed by vortex for 5 s. Based on the calculated protein
  • wCEX. P05 was evaluated by weak cation exchange chromatography (wCEX) using a Dionex ProPac® WCX-10 4x250 mm column (Product Number 054993), with a flow rate of 1 .2 mL/min using mobile phase solutions of 10 mM sodium acetate pH 5.5 (buffer A) and 10 mM sodium acetate pH 5.5, 250 mM NaCI (buffer B). A gradient is performed from 10%B to 25%B over 20 minutes. Intact P05 elutes approximately 1 .5 to 2.5 minutes before the des-Ala species in the later part of the gradient.
  • Wash #2 results in a peak comprised mainly of impurities. Elutions #1 with 30% and 35% B result in >95% pure product by SDS-PAGE analysis. The salt concentration of Wash #2 can be increased by a small amount to remove the shoulder on the elution peak.
  • P04 was purified, and diffraction quality crystals were grown in 25% PEG1500, 0.1 M PCB (pH 4.0) at 20 Q C.
  • the X-ray structure of P04 was solved by molecular replacement using a model incorporating the relevant portions of known IL-1 ⁇ and IL-1 Ra structures from PDB structures 1 1TB and 1 1RA (Vigers et al., (1997) Nature 386: 190-194 and Schreuder et al., (1997) Nature 386: 194-200).
  • the final model was refined to a R wor k Rfree of 17.6%/20.4% and contains one P04 molecule (140 residues) and 98 water molecules (Table 10). P04 residues 1 -2, 48-49 and 85-93 were not visible in the electron density and are missing in the final model.
  • R free was calculated from a subset of reflections (5%) not used for refinement.
  • the P04 crystal structure has a fold similar to that predicted by the modeling.
  • a view of this structure is shown in FIG. 1 .
  • An overlay of the two structures is shown in FIG. 12A.
  • the RMSD for backbone carbons was 1 .41 A and 1 .09 A for the IL-1 ⁇ and IL-1 Ra segments versus the same residues on the respective parent molecules.
  • P04 has at least one unique salt bridge and two unique hydrogen bonds that involve an interaction between a residue derived from IL-1 ⁇ and a counterpart derived from IL-1 Ra: (i) Glu39- Lys64 (Glu39 from IL-1 RA; Lys64 from IL-1 ⁇ ) as shown in FIG.

Abstract

Featured herein are non-naturally occuring cytokine domains that can be used, inter alia, to modulate cellular signalling responsive to interleukin-1 receptor I (IL-1 RI), to treat disorders, and to detect and/or bind to cellular receptors, as well as other agents. Exemplary cytokine domains can contain amino acid residues from at least two parental cytokines domains, for example, receptor binding features, surface features, β strands, and loops from at least two parental cytokines domains.

Description

CHIMERIC IL-1 RECEPTOR TYPE I AGONISTS AND ANTAGONISTS
CROSS-REFERENCE AND CLAIM OF PRIORITY
This application claims priority to United States provisional patent applications Serial No. 61/368,799, filed July 29, 2010, Serial No. 61/436,178, filed January 25, 201 1 , Serial No. 61/436,184, filed January 25, 201 1 , Serial No. 61/493,966, filed June 6, 201 1 , and Serial No. 61/493,967, filed June 6, 201 1 , and with respect to United States applications incorporates the contents of Serial No. 61/368,799, filed July 29, 2010, Serial No. 61/436,184, filed January 25, 201 1 , and Serial No. 61 /493,967, filed June 6, 201 1 , by reference.
BACKGROUND
lnterleukin-1 alpha (IL-1 a) and beta (IL-1 β) are prototypic members of a family of immunoregulatory cytokines and have several prominent roles in regulating the immune system. IL-1 a and IL-1 β bind to the interleukin-1 receptor I (IL-1 Rl), leading to the engagement of the secondary receptor, interleukin-1 receptor accessory protein (IL- 1 RAcP). Signaling agonized by IL-1 a and IL-1 β leads to amplified T cell responses, including the proliferation and survival of na'ive T cells and the development of TH17 cells.
SUMMARY
Featured herein are non-naturally occuring cytokine domains that can be used, inter alia, to modulate cellular signalling responsive to interleukin-1 receptor I (IL-1 Rl), to treat disorders, and to detect and/or bind to cellular receptors, as well as other agents.
In one aspect, this disclosure features an isolated protein including an cytokine domain that contains amino acid residues from at least two parental cytokines domains, for example, receptor binding features, surface features, β strands, and loops from at least two parental cytokines domains.
In some embodiments, the cytokine domain binds to IL-1 Rl and includes receptor binding features from different parental cytokine domains, e.g., from a receptor agonist and a receptor antagonist (such as IL-1 β and IL-1 Ra, or IL-1 a and IL-1 Ra), from IL-1 β and IL-1 a, or from all three of IL-1 Ra, IL-1 a and IL-1 Ra. The receptor binding features can correspond to residues, segments, or regions in Sites A and B. With respect to such residues, segments, and regions corresponding to Sites A and B, in the context of IL-1 (IL-1 β, IL-1 a, and IL-1 Ra), see the definitions further below. With respect to Site A, the cytokine domain may have: (a)(i) Site A residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in a first parental cytokine domain; (a)(ii) Extended Site A residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in a first parental cytokine domain; (a)(iii) Site A segments A1 and A2 that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding regions of a first parental cytokine domain; and/or (a)(iv) a Site A region that is at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding regions of a first parental cytokine domain.
With respect to Site B, the cytokine domain may have: (b)(i) Site B residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in a second parental cytokine domain; (b)(ii) extended Site Bs residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in a second parental cytokine domain; (b)(iii) Site B segments B1 , B2, and B3 that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding regions of a second parental cytokine domain; and/or (b)(iv) a Site B region that is at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding regions of a second parental cytokine domain.
In some embodiments, the cytokine domain includes features: (a)(i) and b(i), (a)(ii) and b(ii), (a)(iii) and (b)(iii), or (a)(iv) and (b)(iv), e.g., wherein each feature is further defined by 80, 85, 88, 90, 92, 95, or 100% identity. For example, the first parental cytokine domain can be I L- 1 β , and the second parental cytokine domain can be IL-1 Ra. For example, the first parental cytokine domain can be IL-1 a, and the second parental cytokine domain can be IL-1 Ra.
The cytokine domain can also include amino acids from a second parental cytokine domain at one or more positions in the domain that impair interaction with a cytokine secondary receptor (e.g., IL-1 RAcP). For example, the second parental cytokine domain is IL-1 Ra. In some embodiments, the cytokine domain includes one or more Site C and/or D segments (e.g., C1 , D1 , D2, D3, D4, and/or D5) from IL-1 Ra, or sequences at least 80, 85, 88, 90, 92, 95, or 100% identical to such segments. For example, the cytokine domain includes (i) a Site C residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra, (ii) Site D residues that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra, (iii) a C1 segment that is at least 70, 75, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra; or (iv) a D2 segment that is identical at at least 3, 4, or 5 residues to corresponding residues in IL- 1 Ra. The cytokine domain can include features (i) and (ii), or (ii) and (iii), e.g., wherein each feature is further defined by 80, 85, 88, 90, 92, 95, or 100% identity, or (iii) and (iv) The domain can include regions from at least two different human IL-1 family cytokine domains, wherein the regions are selected from the group consisting of the A region (having A1 and A2 segments), the B region (having B1 , B2, and B3 segments), the C region, and the D region (having D1 , D2, D3, D4, and D5 segments).
The cytokine domain can include a Site A region and a Site B region from different cytokine domains. The Site A region can be from a naturally occuring receptor agonist or antagonist; the Site B region can be from a naturally occuring receptor agonist. It can include a Site C region from a naturally occuring receptor antagonist and/or a Site D region from a naturally occuring receptor antagonist.
For example, the domain can be a chimeric domain having segments that are at least 5, 6, 10, 15, 20, or 25 amino acids in length and that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding segments from at least two different parental cytokine domains, such as a first and second parental cytokine domain. The parental cytokine domains can be IL-1 Rl binding cytokines, such as IL-1 β, IL-1 a, and IL-1 Ra. In some embodiments, the amino acids that are not in the segments from the first parental cytokine domain are from two or more other parental cytokine domains.
In some embodiments, the cytokine domain includes at least two segments of at least 5, 6, 10, 15, 20, or 25 amino acids in length that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding segments of a first parental cytokine domains, and the amino acids that are not in such segments are predominantly (e.g., at least 50, 60, 70, 80, 85, 88, 90, 92, 95, or 100%) identical to corresponding residues in the second parental cytokine domain.
In some embodiments, the cytokine domain includes (i) at least two segments of at least 5, 6, 10, 15, 20, or 25 amino acids in length that are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding segments of a first parental cytokine domains, and (ii) at least one, two or three segments, e.g., of at least 5, 6, 7, 8, 10, or 15 amino acids in length, that are identical to a second parental cytokine domain.
For example, the cytokine domain can include a first segment of 20-50, 25-50, 30-45, or 30-40 amino acids (e.g., 29, 30, 31 , 32, 33, 34, 35, 36, 37, 38, 39, or 40), and a second segment of 20-45, 20-40, 25-40, or 25-35 amino acids (e.g., 25, 26, 27, 28, 29, 30, 31 , 32, 33, 34, or 35), each identical (or at least 80, 85, 88, 90, 92, 95, or 98% identical) to a first parental cytokine domain (e.g., IL-1 Ra), and a third segment that is identical (or at least 80, 85, 88, 90, 92, 95, or 98% identical) to a second parental cytokine domain (e.g., IL-1 β or IL-1 a). For example, the third segment can be between 55-90, 60-90, 60-85, or 70-85 amino acids in length, e.g., 75, 76, 77, 78, 79, 80, 81 , 82, 83, 84, or 85 amino acids in length.
In some embodiments: the first segment can be a segment at least 80, 85, 88, 90, 92, 95, 98, or 100% to WDVNQKTFYLRNNQLVAGYLQGPNV (SEQ ID NO:9, also termed the A1 segment herein, and corresponding to residues 16-40 of SEQ ID NO:3 and residues 1 1 -36 according to IL-1 β numbering). The second segment can be a segment at least 80, 85, 88, 90, 92, 95, 98, or 100% to residues 120-140 or 120-141 of SEQ ID NO:3 (IL-1 Ra), corresponding to residues 121 -139 or 121 -140 (according to IL- 1 β numbering). The third segment can be at least 80, 85, 88, 90, 92, 95, 98, or 100% to residues 45-100 or 42-120 from IL-1 β (SEQ ID NO:1 ).
In some cases, the first segment can be a segment at least 80, 85, 88, 90, 92, 95, 98, or 100% to residues 14-45 of SEQ ID NO:3 (corresponding to residues 9-41 according to IL-1 β numbering). The second segment can be a segment at least 80, 85, 88, 90, 92, 95, 98, or 100% to residues 120-145 of SEQ ID NO:3 (corresponding to residues 121 -145 according to IL-Ι β numbering) or residues 120-147 of SEQ ID NO:3 (corresponding to residues 121 -147 according to I L- 1 β numbering). In some
embodiments, at least residues 1 1 -41 and 120-147 (according to IL-1 β numbering) are collectively at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra.
In some embodiments, a terminus of one of the segments of the first IL-1 family cytokine domain is located within five, four, three, two, or one amino acids of amino acid 41 of SEQ ID NO:1 and a terminus of one of the segments of the first IL-1 family cytokine domain is located within five, four, three, two, or one amino acids of amino acid 121 of SEQ ID NO:1 .
In some embodiments, the domain includes a segment having an N-terminus at a position within five, four, three, two, or one amino acids of amino acid 42 of SEQ ID NO:1 and a C-terminus within five, four, three, two, or one amino acids of amino acid 120 of SEQ ID NO:1 , and/or a segment having an N-terminus at a position within five, four, three, two, or one amino acids of amino acid 121 of SEQ ID NO:1 and a C-terminus within five, four, three, two, or one amino acids of amino acid 145 of SEQ ID NO:1 . In some embodiments, residues in the domain at positions corresponding to 1 1 -41 and 120-147 (according to IL-1 β numbering) are collectively at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra. The domain can also be based on sequences from IL-1 cytokine family members, e.g., at analogous positions to the foregoing.
In some embodiments, the cytokine domain includes one, two, three or more of the following sequences: WDVNQKTFYLRNNQLVAGYLQGPNV (SEQ ID NO:9);
NLEEK (SEQ ID NO:10); RIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEK(SEQ ID NO:1 1 ); AMEADQP(SEQ ID NO:12); FLCTAMEADQPVSLTNMPDEGVMVTKFY(SEQ ID NO:13); and/or sequences at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to the foregoing. In some embodiments, the cytokine domain includes one, two, three or more of the following sequences: VQGEESNDKI (SEQ ID NO:14); KKKMEKRF (SEQ ID NO:15); and FSMSFVQGEESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKN YPKKKMEKRFVFNKIEINNKLEFES (SEQ ID NO:16); and/or sequences at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to the foregoing.
In some embodiments, one or more or all of the β1 β2, β2β3, β8β9 and β10β1 1 loops are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding loops from an IL-1 antagonist, e.g., IL-1 Ra. In some embodiments, one or more or all of the β4β5, β5β6, β6β7 and β7β8 loops are at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding loops from a different IL-1 family cytokine domain than the human parental cytokine that is most similar to the β1 β2, β2β3, β8β9 and β10β1 1 loops. For example, one or more or all of the β4β5, β5β6, β6β7 and β7β8 loops are at least 80, 85, 88, 90, 92, 95, or 100% identical to such loops from an IL-1 agonist such as I L- 1 β . In some embodiments, the β1 1 β12 loop is at least 80, 85, 88, 90, 92, 95, or 100% identical to corresponding loop from an IL-1 antagonist, e.g., IL-1 Ra.
In some embodiments, the cytokine domain includes sequences that are at least 80, 85, 88, 90, 92, 95, or 100% identical to one, two, three, or all of beta strands β2, β3, β10 and β1 1 of IL-1 Ra. In some embodiments, the cytokine domain includes sequences that are at least 80, 85, 88, 90, 92, 95, or 100% identical to one, two, three, or all of β4, β6, β7, and β8 of IL-1 β, or to β4, β5, β6, β7, and β8 of IL-1 β.
In some embodiments, the cytokine domain does not contain a segment that is greater than 80, 85, 90, 95, or 100% identical to amino acids I46-G59, A55-G59, A55- V83, I60-V83, N84-D95, I46-S1 10, V49-S1 10, or I46-G1 18 of SEQ ID NO:3. In some embodiments, the cytokine domain does not contain a segment that is greater than 80, 85, 90, 95, or 100% identical to amino acids N7-V41 , R1 1 -M36, N102-D145, or Y121 - D145 of SEQ ID NO:1 .
Generally, the cytokine domain is not naturally occurring. It differs from human IL-1 family cytokine domains. For example, it is less than 98, 95, 90, 85, 80, 75, 70, 65, 60, or 55% identical to IL-1 Ra (SEQ ID NO:3), IL-1 β (SEQ ID NO:1 ), and/or IL-1 a (SEQ ID NO:2). The cytokine domain can also be at least 30, 40, 45, 50, 55, 60, 65, 70% identical to such cytokine. For example, the chimeric domain can be between 30-95%, 40-90%, or 45-85% identical to IL-1 Ra, IL-1 β , and IL-1 a. For example, the chimeric domain can be between 40-90% identical to IL-1 β and 35-85% identical to IL-1 Ra;
between 40-80% identical to IL-1 β and 45-80% identical to IL-1 Ra; between 45-72% identical to IL-1 β and 45-80% identical to IL-1 Ra; between 45-72% identical to IL-1 β and 53-80% identical to IL-1 Ra; between 50-72% identical to IL-1 β and 53-70% identical to IL-1 Ra; between 60-72% identical to IL-1 β and 53-68% identical to IL-1 Ra; between 65- 72% identical to IL-1 β and 54-60% identical to IL-1 Ra; or between 68-72% identical to I L- 1 β and 54-57% identical to IL-1 Ra. For example, the chimeric domain can be between 40-90% identical to IL-1 a and 35-85% identical to IL-1 Ra; between 40-80% identical to IL-1 a and 45-80% identical to IL-1 Ra; between 45-72% identical to IL-1 a and 45-80% identical to IL-1 Ra; between 45-72% identical to IL-1 a and 53-80% identical to IL-1 Ra; between 50-72% identical to IL-1 a and 53-70% identical to IL-1 Ra; between 60- 72% identical to IL-1 a and 53-68% identical to IL-1 Ra; between 65-72% identical to IL-1 a and 54-60% identical to IL-1 Ra; or between 68-72% identical to IL-1 a and 54-57% identical to IL-1 Ra.
In some embodiments, the cytokine domain differs from IL-1 Ra, and binds to the receptor while including a Site C and/or Site D characteristic of a naturally occurring receptor antagonist (such as IL-1 Ra). For example, the domain is less than 98, 95, 92, 90, 85, and 80% identical from human IL-1 β and/ IL-1 Ra. For example, the domain is between 40-95%, 40-90% or 45-85% identical to IL-1 Ra. The domain can also be between 40-95%, 40-90% or 45-85% to an IL-1 family cytokine agonist, such as IL-1 a or I L- 1 β . In some embodiments, the cytokine domain includes: at least 40, 45, 50, 55, 60, 65, 70, 75, 80 or 85% amino acids from a first parental cytokine domain that is an agonist of cytokine signaling. In some embodiments, the cytokine domain has greater amino acid identity (e.g., at least 5, 10, 15, or 20% greater) to a receptor agonist (such as IL-1 β or IL-1 a) than to a receptor antagonist (IL-1 Ra), but functions as an antagonist of IL-1 Rl.
In some embodiments, the cytokine domain is completely chimeric, e.g., each amino acid in the domain is from one of the parental cytokine domains, e.g., one of two parental cytokine domains or one of three or more parental cytokine domains. For example, the parental cytokine domains are human cytokine domains or non-human primate cytokine domains. In some embodiments, the cytokine domain is partially chimeric, e.g., not all amino acids in the domain are from one of the parental cytokine domains.
For example, the isolated protein binds to IL-1 Rl and modulates signaling by the receptor, e.g., agonizes or antagonizes IL-1 Rl receptor signaling activity. In some embodiments, the protein does not substantially induce IL-6 production when contacted to IL-1 β responsive human cells and/or does not substantially induce production of an IL-1 β responsive reporter gene, e.g., at concentrations of 10 g ml, 100 g ml, or 1 mg/ml. Generally, the protein inhibits signaling by IL-1 β (e.g., at a concentration of 0.1 ng/ml such as in a cellular assay described herein) with an IC50 of less than 100, 50, 20, 10, or 5 nM. The protein can inhibit signaling by IL-1 β with an IC50 that is less than that of IL-1 Ra, e.g., at least 10, 20, or 50% lower.
In certain embodiments, the cytokine domain binds to e.g., with the same or better affinity than one of the parental cytokine domains. In some embodiments, the cytokine domain binds to IL-1 Rl with KD of less than 100, 50, 20, 10, 5, or 1 nM, or less than 500, 400, 100, or 50 pM. For example, the association constant can be greater than 1 x 1 04, 3χ 1 04, 1 χ 1 05, or 1 χ 106 M"V, and the dissociation can be less than 1 x 10"3, 1 x 1 0"4, 6x 1 0"4, or 6x 1 0"5 S"1 .
In some embodiments, the cytokine domain binds to IL-1 Rl with a better affinity (e.g., lower KD) and/or a slower dissociation rate than IL-1 β or IL-1 Ra. For example, the cytokine domain can bind to IL-1 Rl with a dissociation constant less than or equal to that of IL-1 Ra and/or with an association constant greater or equal to that of IL-1 β.
The cytokine domain can be between approximately 120-180, 140-170, or 148-
160, or 150-156 amino acids in length. In some embodiments, the domain is 152 or 153 amino acids in length. Typically the domain includes at least 10, 1 1 , or 12 β-strands, and is stably folded. In some embodiments, the cytokine domain has a Tm of at least 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, or 64°C, as described herein. It can have a Tm of between 51 -61 , 51 -66, 56-61 , or 56-66°C. In some embodiments, the cytokine domain does not begin unfolding until at least 48, 50, 51 , 55, 57, 58, or 59°C. For example, it has a Tm that is at least within 10°C or 5°C of the Tm of IL-1 Ra and/or I L- 1 β in a physiological buffer. In some embodiments, it is more thermostable than IL-1 Ra and/or IL-1 β in a physiological buffer. For example the domain can have a Tm that is at least 2, 4, 6, 7, or 8°C greater than the Tm of IL-1 Ra and/or IL-1 β in a physiological buffer, e.g., between about 5-12, 5-10, or 7-10°C greater than the Tm of IL-1 Ra and/or IL-1 β at a concentration of about 0.5 mg/ml.
The protein can include other features described herein.
In another aspect, the disclosure features an isolated protein that includes a chimeric IL-1 family cytokine domain. Examples of IL-1 cytokine family members include IL-1 a, IL-1 β, IL-1 Ra, IL-18, IL-1 F5, IL-1 F6, IL-1 F7, IL-1 F8, IL-1 F9, IL-1 F10, and IL-33. The cytokine domain can include a receptor binding region of one of the foregoing cytokines or a protein sequence that includes elements of one or more such cytokines. For example, the cytokine domain can include a chimera of two or more IL-1 cytokine family members.
In one embodiment, the chimeric domain includes at least one segment of length at least five, six, seven, eight, nine, ten, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, or 70 amino acids and having amino acid identity to (or at least 80, 82, 85, 87, 90, 92, 94, 95, 96, 97, 98, or 99% identity to) to a first IL-1 family cytokine, and another segment of length of length at least five, six, seven, eight, nine, ten, 15, 20, 25, 30, 35, or 40 amino acids and having amino acid identity to (or at least 80, 82, 85, 87, 90, 92, 94, 95, 96, 97, 98, or 99% identity to) to a second IL-1 family cytokine. The chimeric domain can be less than 90, 85, 80, or 75% identical to one or both of the first and second IL-1 family cytokine.
In one embodiment, the first and second IL-1 family cytokines are selected from the group consisting of IL-1 β, IL-1 a, and IL-1 Ra. In another embodiment, the first and second IL-1 family cytokines are selected from the group consisting of IL-1 F5, IL-1 F6, IL-1 F7, and IL-1 F8. In another embodiment, the first IL-1 family cytokine is selected from the group of agonists and the second is selected from the group of antagonists. In some embodiments, the chimeric domain includes fewer than 120, 1 10, 100, 90, or 80 contiguous amino acids from the same parental cytokine domain. In one embodiment, the chimeric domain is identical to the first IL-1 family cytokine at at least 50, 60, 70, 80, 90, 100, 1 10, or 120 positions and is identical to the second IL-1 family cytokine at at least 50, 60, 70, 80, 90, 100, 1 10, or 120 positions (including positions that may individually be identical to both such cytokines).
In one embodiment, the chimeric domain includes at least two, three, or four discontinuous segments, each of length at least five, six, seven, eight, nine, ten, 15, or 20 amino acids and having amino acid identity to (or at least 80, 82, 85, 87, 90, 92, 94, 95, 96, 97, 98, or 99% identity to) corresponding segments of a first IL-1 family cytokine and including amino acids predominantly from a second IL-1 family cytokine at remaining positions. In one embodiment, the chimeric domain includes 4, 5, 6, or 7 segments, wherein adjacent segments are from different parental IL-1 family cytokine domains. For example, each amino acid in the domain is located in a peptide of at least 5 or 6 amino acids in length from a naturally occurring human IL-1 family cytokine domain. In one embodiment, the chimeric domain includes at least one, two, or three of: (i) a segment of at least 50, 60, 65, 70, or 75 amino acids in length from IL-1 β , (ii) a segment of at least 15, 20, 25 amino acids in length from IL-1 Ra; and (iii) another segment of at least 15, 20, 25 amino acids in length from IL-1 Ra.
In one embodiment, the discontinuous segments includes residues (i) 1 -6 and 45-61 , (ii) 1 -6 and 86-95, (iii) 45-61 and 86-95, (iv) 1 -6 and 148-153, (v) 45-61 and 148- 153, or (vi) 86-95 and 148-153, according to the numbering of such positions in
IL-1 β. The three discontinuous segments from the first IL-1 family cytokine can include, e.g., residues 1 -8, 42-120, and 141 -153, residues 1 -10, 37-125, and 131 -153, or residues 1 -6, 45-61 , 86-95, and 148-153, according to the numbering of such positions in IL-1 β. The chimeric domain can be at least 80, 82, 85, 87, 90, 92, 94, 95, 96, 97, 98, 99, or 100% identical to the second IL-1 family cytokine at the remaining positions. In one embodiment, one or more of the borders of the discontinuous segments are located at positions wherein the first and second IL-1 family cytokines are identical or are conserved. The protein can have other features described herein.
In another aspect, this disclosure features an isolated IL-1 inhibitor including a IL- 1 family cytokine domain that binds to IL-1 Rl. For example, the IL-1 inhibitor includes one or more features above or elsewhere herein. In some embodiments, the cytokine domain includes: (a) amino acids identical to IL-1 Ra at the following positions ARG1 1 , SER13, GLN14, GLN15, GLU25, LYS27, LEU29, HIS30, LEU31 , GLN32, GLY33, GLN34, ASP35, MET36, GLN38, GLN39, ALA127, GLU128, ASN129, MET130, and GLN141 (according to the numbering of IL-1 β) and (b) amino acids identical to IL-1 β at the following positions: ALA1 , PR02, VAL3, ARG4, LEU6, PHE46, GLN48, GLU51 , SER52, ASN53, LYS55, ILE56, PR057, LYS92, LYS93, LYS94, LYS103, GLU105, ASN108, GLN149, PHE150, and SER152. In some embodiments, Site A segments A1 and A2 are at least 80% identical (collectively) to corresponding segments of IL-1 Ra. In some embodiments, Site B segments B1 , B2, and B3 are at least 80% identical (collectively) to corresponding segments of IL-1 β. For example, Site A segments A1 and A2 are at least 90% identical (collectively) to corresponding segments of IL-1 Ra; and Site B segments B1 , B2, and B3 are at least 90% identical (collectively) to corresponding segments of IL-1 β. For example, Site A segments A1 and A2 are identical to
corresponding segments of IL-1 Ra; and Site B segments B1 , B2, and B3 are identical to corresponding segments of IL-1 β .
In some embodiments, the cytokine domain includes sequences that are at least 80% identical (collectively) to beta strands β2, β3, β10 and β1 1 of IL-1 Ra and sequences that are at least 80% identical (collectively) to beta strands β4, β6, β7, and β8 of IL-1 β. In some embodiments, the cytokine domain includes sequences that are identical to beta strands β2, β3, β10 and β1 1 of IL-1 Ra and sequences that are identical to beta strands β4, β6, β7, and βδ of IL-1 β . In some embodiments, the segments and features identified above from IL-1 β are derived from IL-1 a, or a combination of IL-1 β or IL-1 a.
In some embodiments, the IL-1 inhibitor includes one or more (e.g., at least two, three, four, five, six, or seven) of the following properties: (i) Site A or Site B residues (and/or extended Site A and extended Site B residues) that are at least 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 β , IL-1 a, or IL- 1 Ra; (ii) A1 and A2 segments that are collectively at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra; (iii) B1 , B2, and B3 segments that are collectively at least 80% identical to corresponding residues in IL-1 β or IL-1 a; (iv) a Site A region that is at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to
corresponding residues in IL-1 Ra; (v) a Site B region is at least 80, 85, 88, 90, 92, 95, 98, or 100% to corresponding residues in IL-1 β or IL-1 a; (vi) a Site C and/or Site D residues that are at least 50, 60, 70, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra or IL 36Ra, (vii) a C1 segment that is at least 70, 75, 80, 85, 88, 90, 92, 95, 98, or 100% identical to corresponding residues in IL-1 Ra; (viii) a D2 segment that is identical at at least 3, 4, or 5 residues to corresponding residues in IL-1 Ra. The cytokine domain differs from IL-1 Ra, for example, the cytokine domain is less than 99, 98, 95, 90, 85, 80, 75, 70, 65, 60% identical to IL-1 Ra, and/or at least 30, 40, 45, 50, 55, 60, 65, 70% identical to IL-1 Ra, e.g., between 40-95%, 40-90%, or 45- 85% identical to IL-1 Ra. In some embodiments, the cytokine domain does not contain a segment that is greater than 80, 85, 90, 95, or 100% identical to amino acids I46-G59, A55-G59, A55-V83, I60-V83, N84-D95, I46-S1 10, V49-S1 10, or I46-G1 18 of SEQ ID NO:3. In some embodiments, the cytokine domain does not contain a segment that is greater than 80, 85, 90, 95, or 100% identical to amino acids N7-V41 , R1 1 -M37, N102- D144, or Y121 -D144 of SEQ ID NO:1 .
In some embodiments, the inhibitor binds to IL-1 Rl with KD of less than 100, 50,
20, 10, 5, or 1 nM. In some embodiments, the inhibitor binds to IL-1 Rl with a better affinity (e.g., lower KD) and/or a slower dissociation rate than IL-1 β or IL-1 Ra.
In some embodiments, the inhibitor does not substantially induce IL-6 expression when contacted to I L- 1 β responsive human cells. Generally, the inhibitor inhibits signaling by IL-1 β, e.g., with an IC50 of less than 100, 50, 20, 10, or 5 nM.
In some embodiments, the cytokine domain includes one, two, three or more of the following sequences: WDVNQKTFYLRNNQLVAGYLQGPNV (SEQ ID NO:9);
NLEEK (SEQ ID NO:10); RIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEK(SEQ ID NO:1 1 ); AMEADQP(SEQ ID NO:12); FLCTAMEADQPVSLTNMPDEGVMVTKFY(SEQ ID NO:13); and/or sequences at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to the foregoing. In some embodiments, the cytokine domain includes one, two, three or more of the following sequences: VQGEESNDKI (SEQ ID NO:14); KKKMEKRF (SEQ ID NO:15); and FSMSFVQGEESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKN YPKKKMEKRFVFNKIEINNKLEFES (SEQ ID NO:16); and/or sequences at least 80, 85, 88, 90, 92, 95, 98, or 100% identical to the foregoing. The inhibitor can include other features described herein.
In another aspect, the disclosure provides an isolated protein that includes an amino acid sequence at least 80, 82, 85, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% or 100% identical to a sequence disclosed herein, e.g., a sequence listed in Table 4 or Example 1 , e.g., the amino acid sequence of P01 , P02, P03, P04, P05, P06, or P07, or a sequence in Example 1 , 5, 6, or elsewhere herein. In some embodiments, the amino acid sequence includes at least one substitution, insertion, or deletion. The amino acid sequence can include fewer than 15, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, or 2 non- conservative substitutions or fewer than 15, 12, 1 1 , 10, 9, 8, 7, 6, 5, 4, 3, or 2 total substitutions. The amino acid sequence can include at least 1 , 2, 3, 4, or 5 substitutions, e.g., conservative substitutions.
Also provided are isolated proteins that include a methionine N-terminal to the amino acid sequence of P01 , P02, P03, P04, P05, P06, or P07, or a sequence in Example 1 , 5, 6, or elsewhere herein, and isolated proteins that include the amino acid sequence of P01 , P02, P03, P04, P05, P06, or P07, or a sequence in Example 1 , 5, 6 or elsewhere herein in which the alanine at N-terminus is absent. The foregoing
sequences can include other features disclosed herein. For example, the sequence can further include a tag, such as a hexa-histidine sequence, e.g., N- or C-terminal relative to the IL-1 Rl binding sequence. The sequence can further include a moiety that modifies the stability or pharmacokinetics of the IL-1 Rl binding sequence. For the sequence can further include a serum albumin and/or an Fc domain, or one or more domains thereof, e.g., one or more immunoglobulin constant domains or one or more albumin domains. The protein can have other features described herein. In some embodiments, the isolated protein consists of or consists essentially of a sequence or chimeric domain disclosed herein.
In still another aspect, the disclosure provides an isolated protein that includes a domain having a circularly permuted form of a cytokine domain described herein, e.g., a cytokine domain listed in Table 4 and/or a domain that includes SEQ ID NO:3. The protein can further include a heterologous sequence (such as an Fc domain or albumin) at the N- or C-terminus of the permuted form, optionally spaced by a linker. The protein can have other features described herein.
The disclosure also features pharmaceutical compositions that include one or more receptor binding agents described herein (such as a protein that includes a chimeric cytokine domain). The compositions can be ophthalmic pharmaceutical compositions, topical compositions, or compositions for parenteral administration.
In another aspect, the disclosure features a method of modulating an immune or inflammatory response in a subject. The method can include administering a
composition that includes a receptor binding agent described herein to a subject in an amount effective to modulate the immune or inflammatory response in the subject.
In another aspect, the disclosure features a method of treating an IL-1 mediated disorder in a subject. The method includes administering a composition that includes a protein that can bind to IL-1 Rl, e.g., a receptor binding agent described herein, to the subject. For example, the disorder can be an autoimmune disorder, e.g., rheumatoid arthritis or juvenile chronic arthritis, scleroderma, Sjogren's syndrome, ankylosing spondylitis, Behcet's syndrome, an inflammatory bowel disease, asthma, vasculitis, or psoriasis. The disorder can be a disorder associated with aggregate formation, e.g., hyperuricemia, gout, diabetes (including non-insulin dependent diabetes), Alzheimer's disease, secondary reactive amyloidosis, amyotrophic lateral sclerosis (ALS),
Huntington's disease, or Parkinson's disease. The disorder can also be a CAPS (CIAS1 Associated Periodic Syndromes) disorder or other disorder described herein.
In another aspect, the disclosure features a method of treating an IL-1 mediated ocular disorder in a subject. The method can include administering a composition that includes a protein that can bind to IL-1 Rl, e.g., a receptor binding agent described herein, to the subject. For example, the composition is an ophthalmic composition that is administered topically to an eye of the subject or surrounding region. In one embodiment, the disorder is a dry eye disorder. In some embodiments, the subject does not exhibit manifestations of systemic autoimmune disease. In some embodiments, the subject has Sjogren's syndrome. In some embodiments, the subject has graft-versus- host disease (GVHD). In still other embodiments, the disorder is uveitis.
In still another aspect, the disclosure features a method of inhibiting IL-1 activity. The method includes contacting a receptor binding agent that can bind to IL-1 Rl to cells responsive to IL-1 or to a subject. Generally, the protein is provided in an amount effective to inhibit IL-1 activity associated with the cells or in the subject. The protein can be contacted to cells from a subject ex vivo.
In another aspect, this disclosure features an isolated nucleic acid that includes one or more sequences encoding the proteins described herein or a nucleic acid disclosed herein (e.g., in Table 5), a sequence that hybridizes to such nucleic acid, or that is at 80, 82, 85, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, 99% or 100% identical to such nucleic acid. Exemplary hybridizing sequences can be at least 200, 300, 400, 420, or 450 nucleotides in length, e.g., between 420-480 nucleotides in length. The nucleic acid can also include other features disclosed herein.
Also featured is a recombinant host cell that includes a nucleic acid that includes one or more sequences encoding the proteins described herein and a polypeptide chain thereof. A receptor binding agent can be produced by a method that includes maintaining the host cell under conditions that permit expression of the receptor binding agent, and optionally recovering the receptor binding agent, e.g., from cells or media associated with the host cell. For example, the receptor binding agent can be purified from lysate from the cells. The purified receptor binding agent can be formulated, e.g., with one or more of an excipient, a stabilizer, and a buffer.
Also featured is a method of providing a chimeric protein domain. The method includes identifying at least two parental proteins having a common fold (e.g., a first parental protein and a second parental protein), locating at least two segments within the first parental protein and constructing a nucleic acid that has a sequence encoding a chimeric amino acid sequence that includes the two segments from the first parental protein and residues that are predominantly from the second parental protein at remaining positions. The domain can be a domain that is largely composed of β-sheets, or a domain that is largely composed of a-helices, or a domain that has a combination of such elements. For example, the domain can have the fold of a cytokine. The first and second parental proteins can be related by homology, e.g., between 10-40% amino acid identity. In some embodiments the segments from the first protein are located within a single folded protein domain, and the chimeric amino acid sequence includes a form of the folded protein domain that is non-identical to the corresponding domain in the first and second parental protein. In some embodiments, the two parental protein have different functional properties, and the chimeric domain can have properties of one or both of the parental proteins. In some embodiments, the chimeric domain has one binding interface from the first parental protein, and another binding interface from the second parental protein.
Reference is made herein to various regions, segments, and residues in IL-1 family cytokines in relation to Sites A, B, C, and D. The location of such residues, segments, and regions in the sequence of human IL-1 β (SEQ ID NO:1 ) and
corresponding positions are provided below and in Fig. 1 :
Site A. Site A residues in IL-1 β include: ARG1 1 , SER13, GLN14, GLN15,
SER21 , GLU25, LYS27, LEU29, HIS30, LEU31 , GLN32, GLY33, GLN34, ASP35, MET36, GLU128, ASN129, and MET130, and corresponding residues of other IL1 cytokine family members (referred to herein as "Site A residues"). In certain contexts, particularly in connection with IL-1 β, reference is made to "extended Site A residues" which include Site A residues as well as GLN149, and PHE150, and corresponding residues of other IL1 cytokine family members. In addition, it is possible to define a "Site A region" as shown in Fig. 4, including for example an A1 segment (corresponding in IL- 1 β to 1 1 -36 of SEQ ID NO:1 ) and an A2 segment (corresponding in IL-1 β to 125-131 of SEQ ID NO:1 ), and corresponding segments in other IL1 cytokine family members. Site B. Site B residues in IL-1 β include: ALA1 , PR02, ARG4, GLN48, GLU51 , ASN53, ILE56, LYS92, LYS93, LYS94, LYS103, GLU105, and ASN108, and
corresponding residues of other IL1 cytokine family members (referred to herein as "Site B residues"). In certain contexts, particularly in connection with IL-1 β, reference is made to "extended Site B residues" which include Site B residues as well as PHE46 and
SER152 which are outside the Site B region in Fig. 4. In addition, it is possible to define a "Site B region" as shown in Fig. 4, including for example a B1 segment (corresponding in IL-1 β to 1 -5 of SEQ ID NO:1 ), a B2 segment (corresponding in IL-1 β to 48-56 of SEQ ID NO:1 ), and a B3 segment (corresponding in IL-1 β to 92-98 of SEQ ID NO:1 ), and corresponding segments in other IL1 cytokine family members.
Site C. Site C residues in IL-1 β include: ILE104, ILE106, ASN107, LYS109, GLU1 1 1 , THR137, LYS138, GLY139, GLY140, GLN141 , THR144, and ASP145 and corresponding residues of other IL1 cytokine family members (referred to herein as "Site C residues"). In addition, it is possible to define a "Site C region" as shown in Fig. 4, including for example a C1 segment (corresponding in IL-1 β to 136-145 of SEQ ID NO:1 ), and corresponding segments in other IL1 cytokine family members.
Site D. Site D residues in IL-1 β include: LEU6, THR9, LYS63, GLU64, LYS65, and ASN66 and corresponding residues of other IL1 cytokine family members (referred to herein as "Site D residues"). In addition, it is possible to define a "Site D region" as shown in Fig. 4, including, for example, a D1 segment (corresponding in IL-1 β to 6-9 of SEQ ID NO:1 ), a D2 segment (corresponding in IL-1 β to 37-41 of SEQ ID NO:1 ), and a D3 segment (corresponding in IL-1 β to 63-66 of SEQ ID NO:1 ), a D4 segment
(corresponding in IL-1 β to 86-91 of SEQ ID NO:1 ), and a D5 segment (corresponding in IL-1 β to 150-153 of SEQ ID NO:1 ) and corresponding segments in other IL1 cytokine family members.
Further identification of the location of residues and regions for Sites A, B, C, and D can be found by alignment of the cytokine in question to the sequences shown in FIG. 4.
The amino acid sequence of IL-1 β (human) as referenced herein is:
APVRSLNCTLRDSQQKSLVMSGPYELKALHLQGQDMEQQVVFSMSFVQGEESNDKIP VALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEKRFVFNKIEINNKLEFESAQF PNWYISTSQAENMPVFLGGTKGGQDITDFTMQFVSS (SEQ ID NO:1 ). The amino acid sequence of IL-1 oc (human) as referenced herein is:
SAPFSFLSNVKYNFMRIIKYEFILNDALNQSIIRANDQYLTAAALHNLDEAVKFDMGAYKS SKDDAKITVILRISKTQLYVTAQDEDQPVLLKEMPEIPKTITGSETNLLFFWETHGTKNYF TSVAHPNLFIATKQDYWVCLAGGPPSITDFQILENQA (SEQ ID NO:2).
The amino acid sequence of IL-1 Ra (human) as referenced herein is:
RPSGRKSSKMQAFRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKIDVVPIEPHALFL GIHGGKMCLSCVKSGDETRLQLEAVNITDLSENRKQDKRFAFIRSDSGPTTSFESAACP GWFLCTAMEADQPVSLTNMPDEGVMVTKFYFQED (SEQ ID NO:3). The terms IL-1 β, IL-1 a, and IL-1 Ra as used herein refer to the respective mature proteins.
The β sheets referenced herein and shown in FIG. 4 refer to the following sequences:
Table 1
Figure imgf000018_0001
Calculations of "homology" or "sequence identity" between two sequences (the terms are used interchangeably herein) are performed as follows. The sequences are aligned according to the alignments provided herein, or, in the absence of an appropriate alignment, the optimal alignment determined as the best score using the Needleman and Wunsch algorithm as implemented in the Needle algorithm of the EMBOSS package using a Blosum 62 scoring matrix with a gap penalty of 10, and a gap extend penalty of 1 . See Needleman, S. B. and Wunsch, C. D. (1970) J. Mol. Biol. 48, 443-453; Kruskal, J. B. (1983) An overview of sequence comparison In D. Sankoff and J. B. Kruskal, (ed.), Time warps, string edits and macromolecules: the theory and practice of sequence comparison, pp. 1 -44 Addison Wesley, and tools available from the European Bioinformatics Institute (Cambridge UK) EMBOSS: The European Molecular Biology Open Software Suite (2000), Rice, P. et al., A., Trends in Genetics 16, (6) pp. 276-277 and available online at http://www.ebi.ac.uk/Tools/emboss/ align/index.html and http://emboss.open-bio.Org/wiki/Appdoc:Needle. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid "identity" is equivalent to amino acid or nucleic acid "homology"). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences. To determine collective identity of one sequence of interest to a group of reference sequences, a position is considered to be identical if it is identical to at least one amino acid at a corresponding position in any one or more of the group of reference
sequences. With respect to lists of segments, features, or regions, identity can be calculated collectively for all members of such list to arrive an overall percentage identity.
Provided herein are sequences that are at least 80, 82, 85, 87, 88, 89, 90, 91 , 92, 93, 94, 95, 96, 97, 98, or 99% identical to sequences disclosed herein.
As used herein, the term "corresponding to" is used to designate the position of an amino acid residue in a polypeptide of interest with respect to a reference
polypeptide. In general the position is the one indicated by an alignment provided herein (e.g., Fig. 4).
As used herein, the term "hybridizes under high stringency conditions" describes conditions for hybridization and washing. Guidance for performing hybridization reactions can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N. Y. (1989), 6.3.1 -6.3.6, which is incorporated by reference. Aqueous and nonaqueous methods are described in that reference and either can be used. High stringency hybridization conditions include hybridization in 6X SSC at about 45 °C, followed by one or more washes in 0.2X SSC, 0.1 % SDS at 65^, or substantially similar conditions. Provided herein are isolated nucleic acids that contain sequences that hybridize under high stringency conditions to nucleic acids encoding amino acid sequences disclosed herein and to the nucleic acids disclosed herein, e.g., in Example 1 .
Naturally occurring proteins referenced herein specifically include the human form of such protein, and also forms from other mammalian species. All patents, published patent applications, and published references cited herein are incorporated by reference for all purposes.
BRI EF DESCRI PTION OF THE DRAWI NGS FIG. 1 is a graphic of the structure of P04 as determined from X-ray
crystallographic data. The backbone of residues from IL-1 Ra are is shown in black, and the backbone of residues from IL-1 β is shown in gray.
FIG. 2 depicts three views of a model of the P05 protein bound to the extracellular domain of human IL-1 Rl. In P05, IL-1 Ra residues are depicted in black and IL-1 β residues are depicted in white
FIG. 3 depicts models of chimeric proteins in which IL-1 Ra residues are depicted in black and IL-1 β residues are depicted in white. The model depicts the proteins: P01 (FIG. 3A), P03 (FIG. 3B), P04 (FIG. 3C), P05 (FIG. 3D), P07 (FIG. 3E), and P06 (FIG. 3F).
FIG. 4 provides an alignment of several human IL-1 family cytokines: IL-1 β (SEQ ID NO:1 ), IL-1 oc (SEQ ID NO:2), IL-1 Ra (SEQ ID NO:3), IL-33 (SEQ ID NO:4), IL-36Ra (SEQ ID NO:5), IL-36oc (SEQ ID NO:6), Ιί-36β (SEQ ID NO:7), and IL-36y (SEQ ID NO:8). Segments referenced in the text herein are identified under the alignment. In addition, β-sheets and loops between such sheets are identified.
FIG. 5A is a listing of the amino acid sequence of P01 (SEQ ID NO:17). FIG. 5B is a listing of the amino acid sequence of P02 (SEQ ID NO:18). FIG. 5C is a listing of the amino acid sequence of P03 (SEQ ID NO:19). FIG. 5D is a listing of the amino acid sequence of P04 (SEQ ID NO:20). FIG. 5E is a listing of the amino acid sequence of P05 (SEQ ID NO:21 ). Segments from IL-1 β are shown in bold italics. See also
Example 1 below.
FIG. 6 is an image of an SDS-PAGE gel showing exemplary samples of protein purified from E. coli cells expressing receptor binding agents. The 15 and 20 kDa molecular weight markers are indicated at left. Lanes are as follows: molecular weight marker (lane 1 and 6), extract (lanes 2 and 7), material purified by cation exchange chromatography (lanes 3 and 8), material additionally purified by anion exchange chromatography (lanes 4 and 9), and reduced samples of such material (lanes 5 and 10). Lanes 2-5 are of P05 purification, and Lanes 6-10 are of P04 purification. See also Example 2. FIG. 7 A is a table and accompanying bar graph showing the ability of the P06, P07, and P01 proteins to agonize signaling relative to IL-1 β and a negative control, β- glucuronidase (GUS) protein. FIG. 7B is a graph depicting antagonism of IL-1 β at various I L-1 β concentrations by P01 .
FIG. 8A is a graph depicting IL-1 β antagonism by P03 (hexa-histidine tagged),
P04 (hexa-histidine tagged), P05 (hexa-histidine tagged), and IL-1 Ra in the presence of 0.1 ng/ml IL-1 β (human). FIG. 8B is a graph depicting IL-1 β antagonism by lysates containing untagged forms of P01 , P02, P03, P04, and P05, and IL-1 Ra in the presence of 0.1 ng/ml IL-1 β (human) and using estimates of the concentration of protein in the respective lysates.
FIG. 9 contains graphs of SPR data showing binding kinetics to immobilized soluble IL-1 Rl for the following proteins: IL-1 β (FIG. 9A), IL-1 Ra (FIG. 9B), P04 (FIG. 9C), and P05 (FIG. 9D).
FIG. 1 0A is a graph depicting thermal denaturation of IL-1 Ra, IL-1 β , P03, P04, and P05 as described in Example 7. FIG. 10B depicts the negative first derivative of the graph in FIG. 10A.
FIG. 1 1 A is a bar graph showing the mean corneal staining score ± SEM as tested by fluorescein staining of the cornea per eye of two independent studies, on days 0, 3, 7, 9, and 1 1 for mice in a dry eye model. The mice received no treatment (n = 18), 10 mg/ml P05 (n=19), or 1 .25χ PBS, the vehicle (n = 20). Asterisks indicate statistical significance of P05 relative to vehicle as follows: * (P < 0.05) and ** (P < 0.005).
FIG. 1 1 B is a bar graph representing data from a separate experiment showing mean corneal staining score ± SEM of the cornea per eye, on days 0, 3, 7, 9, and 1 1 for mice in a dry eye model. The mice received no treatment (n = 8), 1 .25χ PBS vehicle (n =8), 10 mg/ml murine serum albumin (MSA) (n = 8), or 10 mg/ml P05 (n=9). Asterisks indicate statistical significance of P05 relative to murine serum albumin as follows: * (P < 0.05) and *** (P < 0.0005).
FIG. 1 1 C is a bar graph including data for mice that were treated with Restasis® (0.05% cyclosporine emulsion) (n = 8) in the same experiment as FIG. 1 1 B. Asterisks indicate statistical significance of P05 relative to Restasis® as follows: ** (P < 0.005) and *** (P < 0.0005).
FIG. 12A depicts the structure of the X-ray crystallographic structure (black) of P04 overlaid on a computed model (gray) of its structure. FIG. 12B illustrates interactions between K64 and E39 of P04; FIG. 12C illustrates interactions between the C-terminal residues Q149 and S152 with K40 and R9 of P04.
DETAI LED DESCRI PTION
The IL-1 family of cytokines includes several members, all having a common β- trefoil fold comprised of six β-strands that form a β-barrel capped by another six β- strands. The primary structures of the human and other mammalian forms of these cytokines are known. An exemplary structural alignment of several human IL-1 family members is shown in Fig. 1 .
We have discovered, inter alia, that the IL-1 fold is highly plastic. In particular, elements from different members can be combined to provide proteins that agonize or antagonize cytokine signaling. Examples of these proteins include chimeric cytokine domains that include, for example, two or more segments or surface residues from one cytokine in the context of another cytokine or a cytokine consensus sequence, thus creating non-naturally occurring combinations of receptor interaction sites from different IL-1 family cytokines.
IL-1 family cytokines can include at least two primary receptor interaction sites, referred to as Site A and Site B. Sites A and B are involved in contacts to the primary cytokine receptor (e.g., IL-1 Rl). In the case of IL-1 Ra and IL-1 β, for example, the two proteins differ substantially with respect to sites A and B such that IL-1 Ra makes fewer receptor contacts in Site B than Site A.
We have found that it is possible to construct functional chimeric cytokine domains that include a Site A derived from one cytokine and a Site B derived from another cytokine. The plasticity of the IL-1 fold permits construction of a variety of chimeric cytokine domains to antagonize IL-1 signaling.
In addition, IL-1 family cytokines can include two secondary receptor interaction sites, referred to as Site C and Site D, which are involved in agonism and/or antagonism, and can be determinative of a cytokine's ability to interact with its secondary cytokine receptor (e.g., IL-1 RAcP). Inclusion of Site C and/or Site D residues from natural receptor antagonists (such as IL-1 Ra) can impart antagonistic properties. Chimeric cytokine domains can be constructed that include one or both of Sites A and B from one or more IL-1 agonists (such as IL-1 β and IL-1 a) and/or an IL-1 receptor antagonist (such as IL-1 Ra) and one or both of Sites C and D from an IL-1 receptor antagonist (such as IL-1 Ra). Accordingly, it is possible to produce a chimeric cytokine domain that antagonizes signaling and that includes Site B residues from an IL-1 agonist.
Exemplary combinations are also provided in Table 2 below:
Figure imgf000023_0001
The source sequences can be identical to the human sequences for the identified cytokine domains or can contain mutations relative to the human sequences, e.g., such that they are at least 70%, 75%, 80%, 85%, 90%, 95% or more identical to the human sequences in each respective region, e.g., they can include one or more segments from each region.
Sources for Site A residues and Site B residues can be chosen to maximize affinity for the primary receptor. For example, to bind IL-1 Rl, Site A residues can be derived from IL-1 Ra, and Site B residues can be derived from IL-1 β.
A chimeric cytokine domain can have the ability to bind an IL-1 family receptor, e.g., human IL-1 Rl with a KD of less than 10"8, 10"9, or 10"10, e.g., a KD within 10 or 100 fold that of a natural receptor ligand (e.g., I L- 1 β , IL-1 a, or IL-1 Ra) under the same conditions or less than that of a natural ligand (e.g., I L- 1 β , IL-1 a, or IL-1 Ra) under the same conditions. Moreover, in certain embodiments, the chimeric cytokine domain binds with a KD less than, a Kon faster than, or a Koff slower than at least one of its parental cytokine domains.
Chimeric cytokine domains that bind to IL-1 family receptor and which antagonize receptor signaling can be used as receptor binding agents, e.g., to treat disorders mediated by IL-1 family cytokine signaling as described below. For example, in some embodiments, the chimeric cytokine domain binds to IL-1 Rl and antagonizes IL-1 signaling. For example, it has an IC50 of less than 100, 10, 1 , 0.6, or 0.3 nM.
In certain embodiments, the cytokine domain can be at least 40, 45, or 50% identical, but less than completely identical, e.g., less than 95, 90, 85, or 80% identical to a first IL-1 family cytokine domain. At the same time, the cytokine domain can also be at least 40, 45, or 50% identical, but less than completely identical, e.g., less than 95, 90, 85, or 80% identical to a second IL-1 family cytokine domain. The first and second IL-1 family cytokine domain can be less than 50% identical to each other. For example, the first IL-1 family cytokine domain can be an agonist (e.g., IL-1 β or IL-1 a), whereas the second IL-1 family cytokine domain can be a receptor antagonist (e.g., IL-1 Ra).
In some embodiments, at least 80, 85, 90, 92, 94, 95, 97, 98, 99, or 100% of the positions within the cytokine domain have the property that, at each such position, the amino acid present is either identical to the first IL-1 family cytokine domain or to the second IL-1 family cytokine domain (or both if the first and second IL-1 family cytokine are identical at the particular position). Where 100% of the amino acid positions in the cytokine domain have this property, the domain is a complete chimera of two cytokines. Chimeric cytokine domains can also be made from more than two cytokines and can also have mutations relative to its parental cytokines (e.g., one or more particular positions where the amino acid present differs from the corresponding amino acid in each of its parental cytokines).
Cytokine domains can have Site A, B, C, and D residues from different IL-1 cytokine domains, and likewise can have Site A, B, C, and D regions from different IL-1 cytokine domains.
SITE A. For example, in certain embodiments, a cytokine domain
includes residues from a receptor antagonist (e.g., IL-1 Ra) or an agonist at at least 5, 10, 12, 15, 16, 17, or 18 of the Site A residues identified above, or at at least 5, 10, 12, 15, 16, 17, 18, 19, or 20 of the extended Site A residues identified above, or
conservative substitutions of such residues, or at least 50, 65, 75, 80, 90, 95, or 100% of such residues. In some embodiments, the cytokine domain includes residues that are at least 70, 75, 80, 85, 88, 90, 92, 95, or 100% identical to segments A1 , A2, or A1 +A2 in a receptor antagonist (e.g., IL-1 Ra) or an agonist (e.g., IL-1 β or IL-1 a).
In certain embodiments, a cytokine domain includes residues identical to an IL-1 agonist (e.g., IL-1 β residues) at at least 5, 10, 12, 15, 16, 17, or 18 of the Site A residues identified herein, or at least 5, 10, 12, 15, 16, 17, or 18 of the extended Site A residues identified above, or conservative substitutions of such residues, or at least 50, 65, 75, 80, 90, 95, or 100% of such residues.
SITE B. In certain embodiments, a cytokine domain includes residues identical to an IL-1 agonist (e.g., IL-1 β or IL-1 a residues) at at least 2, 3, 5, 8, 9, 10, 1 1 , 12, 13, 14, or 15 of the Site B residues identified herein, or conservative substitutions of such residues, or at least 50, 65, 75, 80, 90, 95, or 100% of such residues. In some embodiments, the cytokine domain includes residues that are at least 70, 75, 80, 85, 88, 90, 92, 95, or 100% identical to segments B1 , B2, B3, B1 +B2, B1 +B3, B2+B3, or B1 +B2+B3 in an IL-1 cytokine agonist (e.g., IL-1 β or ll_-1 oc).
SITE C. In certain embodiments, a cytokine domain includes residues identical to a receptor antagonist (e.g., IL-1 Ra residues) at at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 or 12 of the Site C residues identified herein, or conservative substitutions of such residues, or at least 50, 65, 75, 80, 90, or 100% of such residues. In some embodiments, the cytokine domain includes residues that are at least 50, 65, 75, 80, 90, or 100% identical to the segment C1 in a receptor antagonist (e.g., IL-1 Ra).
In certain embodiments, the cytokine domain can include, for example, one or more of: a hydrophobic amino acid (e.g., Met or lie) at the position corresponding to THR137 of SEQ ID NO:1 , or a hydrophobic, e.g., an aliphatic amino acid (e.g., Val or lie), at the position corresponding to GLN141 of SEQ ID NO:1 , and a non-acidic amino acid, such as a basic amino acid (e.g., Lys or Arg) at the position corresponding to
ASP145 of SEQ ID NO:1 , and such residues at positions corresponding thereto in other IL-1 cytokines. Evidence indicates that Asp145 is important to recruitment of IL-1 RAcP, and accordingly mutation to a non-acidic residue disrupts agonist activity and can be used to confer antagonistic properties. Accordingly, in certain embodiments, a non- acidic amino acid, such as a basic amino acid (e.g., Lys or Arg) or a hydrophobic amino acid, is located at the position corresponding to ASP145 of SEQ ID NO:1 .
SITE D. In certain embodiments, a cytokine domain includes residues identical to a receptor antagonist (e.g., IL-1 Ra residues) at at least one, two, three, four, five, or six of the Site D residues identified herein, or a conservative substitution of such residue, or at least 50, 65, 75, 80, 90, 95, or 100% of such residues. In some embodiments, the cytokine domain includes residues that are at least 50, 65, 75, 80, 90, 95, or 100% identical to segments D1 , D2, D3, D4, D5, D1 +D2, D1 +D2+D3, and combinations thereof in a receptor antagonist (e.g., IL-1 Ra).
Several residues in IL-1 β contact or are in proximity with IL-1 Rl, for example: ALA1 , PR02, VAL3, ARG4, LEU6, ARG1 1 , SER13, GLN14, GLN15, GLU25, LYS27, LEU29, HIS30, LEU31 , GLN32, GLY33, GLN34, ASP35, MET36, GLN38, GLN39, PHE46, GLN48, GLU51 , SER52, ASN53, LYS55, ILE56, PR057, LYS92, LYS93, LYS94, LYS103, GLU105, ASN108, ALA127, GLU128, ASN129, MET130, GLN141 , GLN149, PHE150, and SER152. In addition to the designation into sites as described above, these residues can be classified into two sets: Set 1 and Set 2.
Exemplary Set 1 residues in IL-1 β include: ARG1 1 , SER13, GLN14, GLN15, GLU25, LYS27, LEU29, HIS30, LEU31 , GLN32, GLY33, GLN34, ASP35, MET36, GLN38, GLN39, ALA127, GLU128, ASN129, MET130, and GLN141 and corresponding residues in other IL-1 cytokine family members. Extended Set 1 residues include interaction Set 1 residues and residues within 4 Angstroms of the foregoing in the 1 1TB structure. Exemplary Set 2 residues in IL-1 β include: ALA1 , PR02, VAL3, ARG4, LEU6, PHE46, GLN48, GLU51 , SER52, ASN53, LYS55, ILE56, PR057, LYS92, LYS93, LYS94, LYS103, GLU105, ASN108, GLN149, PHE150, and SER152 and corresponding residues in other IL-1 cytokine family members. Extended Set 2 residues include interaction Set 2 residues and residues within 4 Angstroms of the foregoing in the 1 1TB structure. In certain embodiments, a cytokine domain includes IL-1 β residues at at least 15, 16, 17, 18, 19, 20, or 21 of the Set 1 residues identified above. In certain
embodiments, a cytokine domain includes IL-1 β residues at at least 15, 16, 17, 18, 19, 20, 21 , or 22 of the Set 2 residues identified above. In some embodiments, a cytokine domain includes IL-1 β residues at at least 15, 16, 17, 18, 19, 20, or 21 of the extended Set 1 residues. In some embodiments, a cytokine domain includes IL-1 β residues at at least 15, 16, 17, 18, 19, 20, or 21 of the extended Set 2 residues.
Other variants that can be used as a receptor binding agent include proteins having sequences derived from two or more IL-1 cytokine family members. Examples of such variants include chimeric domains based on IL-1 β and IL-1 Ra. For example, the variants can include one or more amino acid residues from Set 1 of IL-1 Ra (e.g., all Set 1 residues from IL-1 Ra) and one or more amino acid residues from Set 2 of IL-1 β (e.g., all Set 2 residues from IL-1 β).
Exemplary chimeric proteins are predominantly (e.g., at least 50, 60, 70, 75, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 97, 98, 99, or 100%) identical to IL-1 β at the following amino acids positions (i.e., based on correspondence to these positions in IL-1 β):
residues 1 -8, 42-120, and 141 -153 of SEQ ID NO:1 ; residues 1 -6, 45-61 , 86-95, and 148-153 of SEQ ID NO:1 ; and residues 1 -10, 37-125, and 131 -153 of SEQ ID NO:1 .
The remaining residues can be predominantly (e.g., at least 50, 60, 70, 75, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 97, 98, 99, or 100%) identical to IL-1 Ra. For example, the following amino acid positions can be predominantly identical to IL-1 Ra: residues 9-41 and 121 -140 of SEQ ID NO:1 ; residues 7-44, 62-85, and 96-147 of SEQ ID N01 ; and residues 1 1 -36 and 126-130 of SEQ ID NO:1 .
In certain embodiments, the cytokine domain is identical to IL-1 β at at least 2, 4, 5, 10, or 20 positions in addition to amino acid positions Gln48 - Asn53 of IL-1 β, and, e.g., the cytokine domain is predominantly, e.g., at least 50, 60, 70, 75, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 97, 98, 99, or 100% identical to a cytokine other than I L- 1 β .
In certain embodiments, the cytokine domain includes at least 3, 4, 5, 6, 7, or 8 residues identical to IL-1 β at positions corresponding to 1 -8 of SEQ ID NO:1 . For example, it includes residues identical to IL-1 β at positions corresponding to 3, 4, or all 5 of: ALA1 , PR02, VAL3, ARG4, and LEU6.
In certain embodiments, the cytokine domain includes at least 8, 9, 10, 1 1 , 12, 13, 14, 15, 16, or 17 residues identical to IL-1 β at positions corresponding to 45-61 of SEQ ID NO:1 . For example, it includes residues identical to IL-1 β at positions corresponding to 3, 4, 5, 6, 7 or 8 of: PHE46, GLN48, GLU51 , SER52, ASN53, LYS55, ILE56, and PR057.
In certain embodiments, the cytokine domain includes at least 5, 6, 7, 8, 9, or 10 residues identical to IL-1 β at positions corresponding to 86-95 of SEQ ID NO:1 . For example, it includes residues identical to IL-1 β at positions corresponding to one, two, or all three of LYS92, LYS93, and LYS94.
In certain embodiments, the cytokine domain includes at least 3, 4, 5, or 6 residues identical to IL-1 β at positions corresponding to 148-153 of SEQ ID NO:1 . For example, it includes residues identical to IL-1 β at positions corresponding to one, two, or all three of GLN149, PHE150, and SER152.
In certain embodiments, the cytokine domain does not include a threonine at the position corresponding to THR147 in IL-1 β . For example, this position can be an aromatic, such as a tyrosine. The aromatic at the position corresponding to THR147 in IL-1 β can pack against another aromatic (e.g., tryptophan) present in several embodiments at position 1 1 (according to IL-1 β numbering).
In certain embodiments, the cytokine domain does not include an aromatic at the position corresponding to CYS8 in IL-1 β . For example, this position can be an amino acid other than phenylalanine, or other than an aromatic. For example, it can be cysteine, valine, serine, threonine or alanine. This position is located near other bulky residues, particularly MET44 and MET148 (according to I L-1 β numbering). Preferably, not all three of positions 8, 43, and 148 are bulky residues.
In certain embodiments, the cytokine domain includes at least five, six, or seven residues identical to IL-1 Ra at positions corresponding to 30-36 of SEQ ID NO:1 , e.g., at least five, six, or seven residues identical to YLQGPNV (SEQ ID NO:43). For example, the cytokine domain includes at least 10, 12, 14, 16, 18, 19, 20, 21 , 22, 23, 24, 25, or 26 residues identical to IL-1 Ra at positions corresponding to 1 1 -36 of SEQ ID NO:1 . The cytokine domain can also include a basic residue at the position corresponding to 145 of SEQ ID NO:1 .
In certain embodiments, the cytokine domain includes: (a) glutamic acid at the position corresponding to Val40 of IL-1 β and lysine at the position corresponding to Lys65 of IL-1 β ; (b) arginine at the position corresponding to Thr9 of IL-1 β and glutamine at the position corresponding to Gln149 of IL-1 β ; and/or (c) lysine at the position corresponding to V41 of IL-1 β and serine at the position corresponding to Ser152 of IL-1 β.
The cytokine domain can also include at least four, five, six, or seven residues identical to IL-1 Ra at positions corresponding to 126-132, e.g., at least four, five, six, or seven residues identical to MEADQPVS (SEQ ID NO:44).
In certain embodiments, the cytokine domain includes at least 10, 12, 14, 16, 18, 19, 20, 21 , 22, 23, or 24 residues identical to IL-1 β at positions corresponding to 62-85 of SEQ ID NO:1 .
In some embodiments, the cytokine domain includes one or more of the following amino acid sequences (e.g., all four of): RSLAFR (SEQ ID NO:45), IDVSFV (SEQ ID NO:46), KKMDKR (SEQ ID NO:47), and KFYMQF (SEQ ID NO:48); one or more of the following (e.g., all four of) RSLAFR (SEQ ID NO:45), IDVSFV (SEQ ID NO:46), NKLSFE (SEQ ID NO:49), and KFYMQF (SEQ ID NO:48); one or more of the following (e.g., all four of) RSLAFR (SEQ ID NO:45), EEKFSM (SEQ ID NO:50), RFVFIR (SEQ ID NO:51 ), and VTKFTM (SEQ ID NO:52); one or more of the following (e.g., all four of) RSLAFR (SEQ ID NO:45), EEKFSM (SEQ ID NO:50), FESAAC (SEQ ID NO:53), and VTKFTM (SEQ ID NO:54); or one or more of the following (e.g., all four of) LNCRIW (SEQ ID NO:55), EEKFSM (SEQ ID NO:50), PNWFLC (SEQ ID NO:56), and KFYMQF (SEQ ID NO:48).
Chimeric proteins can be used for a variety of purposes. For example, they can be used to increase or decrease receptor signaling activity, to detect cells expressing receptors, or to purify cells or proteins to which they bind. Specific examples of chimeric cytokine domains that are based on IL-1 β and IL-Ra are provided in Example 1 below and include the following exemplary cytokine domains that antagonize IL-1 signaling:
P01 . The P01 domain includes three segments from IL-1 Ra corresponding to amino acids Ala12-Val48, Ne60-Val83, and Asp95-Tyr147 of SEQ ID NO:3, and the remaining four segments from IL-1 β . Overall, the P01 domain has 74 of 153 amino acids from IL-1 β (about 48% identity) and 1 19 amino acids from IL-1 Ra (about 77% identity). These percentages add up to greater than 100% because a number of amino acids in P01 and other exemplary proteins disclosed herein are amino acids that are conserved between IL-1 β and IL-1 Ra and accordingly contribute to the percentage identity for both IL-1 β and IL-1 Ra.
P02. The P02 domain includes three segments from IL-1 Ra corresponding to amino acids Ala12-Val48, Ile60-Val83, and Ser1 10-Tyr147 of SEQ ID NO:3, and the remaining four segments from IL-1 β . Overall, the P02 domain has 85 of 153 amino acids from IL-1 β (about 55% identity) and 108 amino acids from IL-1 Ra (about 70% identity).
P03. The P03 domain includes two segments from IL-1 Ra corresponding to amino acids Ala12-Lys45 and Phe100-Lys145 of SEQ ID NO:3, and the remaining three segments from IL-1 β . Overall, the P03 domain has 94 of 153 amino acids from I L- 1 β (about 61 % identity) and 91 amino acids from IL-1 Ra (about 64% identity).
P04. The P04 domain includes two segments from IL-1 Ra corresponding to amino acids Ala12-Lys45 and Ala1 14-Lys145 of SEQ ID NO:3, and the remaining three segments from IL-1 β . Overall, the P04 domain has 104 of 153 amino acids from IL-1 β (about 68% identity) and 89 amino acids from IL-1 Ra (about 58% identity).
P05. The P05 domain includes two segments from IL-1 Ra corresponding to amino acids Arg14-Lys45 and Phe120-Tyr147 of SEQ ID NO:3, and the remaining three segments from IL-1 β . Overall, the P05 domain has 108 of 153 amino acids from IL-1 β (about 70% identity) and 85 amino acids from IL-1 Ra (about 55% identity).
Other chimeric proteins can be similarly constructed. For example, chimeric proteins can be made between IL-1 a and IL-1 Ra, e.g., specifically domains including the identified segments of IL-1 Ra in combination with corresponding residues from IL-1 a rather than IL-1 β for each of the above examples. Protein Modifications and Substitutions
Protein sequences, such as those described herein, can be varied, e.g., by making one or more conservative substitutions. Conservative substitutions can be made to retain function or to make modest changes in function. Exemplary conservative substitutions are described in the following table:
Table 3
Figure imgf000030_0001
Substitutions can be chosen based on their potential effect on (a) backbone structure in the vicinity of the substitution, for example, a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the volume and branching of the side chain. Amino acid residues can be classified based on side-chain properties: (1 ) hydrophobic: norleucine, met, ala, val, leu, ile;(2) neutral hydrophilic: cys, ser, thr; asn; gin; (3) acidic: asp, glu; (4) basic: his, lys, arg; (5) residues that affect backbone conformation: gly, pro; and (6) aromatic: trp, tyr, phe.
Non-conservative substitutions can include substituting a member of one of these classes for a member of a different class. Conservative substitutions can include substituting a member of one of these classes for another member of the same class. The significance of a particular residue can also be evaluated in the context of the hydropathic index for the amino acid. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics:
isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine (+2.5); methionine (+1 .9); alanine (+1 .8); glycine (-0.4); threonine (-0.7); serine (-0.8);
tryptophan (-0.9); tyrosine (-1 .3); proline (-1 .6); histidine (-3.2); glutamate (-3.5);
glutamine (-3.5); aspartate (-3.5); asparagine (-3.5); lysine (-3.9); and arginine (-4.5). For a discussion of the hydropathic amino acid index and its significance see, for example, Kyte et al., 1982, J. Mol. Biol. 157:105-131 .
The sequence of a protein can be varied by any method, including oligonucleo- tide-mediated (site-directed) mutagenesis, (Carter et al., Nucl. Acids Res. ,13:4331 , 1986; Zoller et al., Nucl. Acids Res., 10:6487, 1987), cassette mutagenesis (Wells et al., Gene, 34:315, 1985), restriction selection mutagenesis (Wells et al., Philos. Trans. R. Soc. London, 317:415, 1986), and PCR mutagenesis. See also In Vitro Mutagenesis Protocols: Third Edition, Braman (ed.), Humana Press, (2010) ISBN: 1607616513 and PCR Cloning Protocols: From Molecular Cloning to Genetic Engineering, Chen and Janes (ed.), Humana Press, (2002) ISBN: 0896039730.
Scanning amino acid analysis can be employed to evaluate one or more amino acids along a contiguous sequence. The method can involve mutating each or nearly each amino acid in a region to a particular amino acid, e.g., to a relatively small, neutral amino acid such as alanine, serine, or valine. Alanine is typically chosen because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main- chain conformation of the variant (Cunningham and Wells, Science, 244: 1081 -1085, 1989). It is also the most common amino acid and is frequently found in both buried and exposed positions (Creighton, The Proteins, (W. H. Freeman & Co., N.Y.); Chothia, J. Mol. Biol., 150:1 , 1976). The scanning process can also be adapted to make more marked changes, e.g., charged residues can be changed to residues of the opposite charge, residues with short side chains can be replaced with ones with bulk side chains. For example, arginine scanning is an approach that can be used instead of or in addition to alanine scanning. The scanning can be applied to each residue in a region or to residues of a particular property, e.g., residues at or near the surface of a protein or likely to be at or near the surface of the protein.
The structure of a protein, one of its domains, or a complex involving the protein can be modeled, e.g., by performing homology based modeling, energy minimization and/or other modeling using known solved structures. Such methods include: Accelrys Software Inc., Discovery Studio®, Release 3.0 , San Diego: Accelrys Software Inc., 2010, AMBER™ modeling software (Case et al. (2005) J. Computat. Chem. 26, 1668- 1688 and Case et al. (2010), AMBER 1 1 , University of California, San Francisco, CA USA) and CHARMM™ modeling software (Molecular Simulations Inc.). See also generally Baker and Sali, Science 294(5540):93-6, 2001 ). The structure can also be determined directly, e.g., using X-ray crystallography and/or NMR spectroscopy.
Exemplary PDB structures describing the structure of IL-1 family cytokines include: 1 11 B, 1 1LR, 1 1RA, 1 1TB, 211 B, 2ILA, 2KLL, 411 B, 511 B, 611 B, 711 B, 811 B, 9ILB, and 1 MD6 (available by http from www.pdb.org from RCSB-Rutgers, Piscataway NJ, USA, and from the National Library of Medicine (Bethesda, MD, USA)). For example, the structures of IL-1 β alone and in complex with the receptor IL-1 Rl have been solved. See e.g., Finzel et al. (1989) J. Mol. Biol. 209: 779-791 , PDB 1 1TB, and Vigers et al. (1997) Nature 386: 190-194. The structure of IL-1 Ra with IL-1 RI has also been solved. See, e.g., PDB 1 1RA and Schreuder et al., (1997) Nature 386: 194-200.
Homology modeling can be assisted by alignment of sequences, e.g., using computer software such as Basic Local Alignment Search Tool (BLAST), PSI-BLAST, PHI-BLAST, WU-BLAST-2, and/or MEGABLAST. See Altschul et al., 1990, J. Mol. Biol. 215, 403-410; Altschul et al., 1996, Methods in Enzymology 266, 460-480; and Karlin et al., 1993, PNAS USA 90, 5873-5787. Additional algorithms for aligning macromolecules (amino acid sequences and nucleic acid sequences) include FASTA (Pearson, 1995, Protein Science 4, 1 145-1 160), ClustalW (Higgin et al., 1996, Methods Enzymol. 266, 383-402), DbClustal (Thompson et al., 2000, Nucl. Acids Res. 28, 2910-2926), and the Molecular Operating Environment (Chemical Computing Group, Montreal, Quebec Canada H3A 2R7). In addition, the algorithm of Myers and Miller (Myers & Miller,
CABIOS 4, 1 1 -17, 1988) which is incorporated into the ALIGN program (version 2.0) of the GCG sequence alignment software package can be used.
A consensus sequence for IL-1 β and IL-1 Ra can be obtained by comparing the two sequences and identifying residues that are identical or that are highly conserved. A chimeric cytokine domain described herein can have at least 60, 70, 80, 90, 95, or 100% of such identical residues or highly conserved residues. An exemplary consensus sequences is: DXXQKX{8_9}L-AXXLQGX{18-28}LGX{7}LSCVXXXDXXXLQLEXVX{8_ 9}KXXKRFXFX{10}FESAXXPXWXXXTXXXXXXPVXLX{5-6}GXXXTXFXXQ (SEQ ID NO:57), where X is independently any amino acid and the subscripted number or range is the number of occurrences. A chimeric cytokine domain described herein can have at least 60, 70, 80, 90, 95, or 100% identity to the consensus sequence (wherein X residues are not counted towards identity). Other consensus sequences can be identified in like manner.
Further variants of an IL-1 cytokine family member can be made and evaluated using a display-based system or other library based screening. For example, the protein variants can be displayed or expressed and evaluated for ability to bind to a receptor for the IL-1 cytokine family member. For example, variants of IL-1 β, IL-1 Ra, or a cytokine domain described herein can be evaluated for ability to bind to the soluble extracellular domain of IL-1 Rl. A general description of display-based systems include the following: for cell display, Chao et al. Nat Protoc. 2006;1 (2):755-68; Colby et al. Methods
Enzymol. 2004;388:348-58; Boder et al., Methods Enzymol. 2000;328:430-44), for phage display (e.g., Viti et al., Methods Enzymol. 2000;326:480-505 and Smith (1985) Science 228:1315-1317), and for ribosome display (e.g., Mattheakis et al. (1994) Proc. Natl. Acad. Sci. USA 91 :9022 and Hanes et al. (2000) Nat Biotechnol. 18:1287-92; Hanes et al. (2000) Methods Enzymol. 328:404-30; and Schaffitzel et al. (1999) J Immunol Methods. 231 (1 -2):1 19-3.
Although many embodiments herein are exemplified using human IL-1 cytokine sequences as parental domains, other sequences can be used. Numerous other IL-1 cytokines, e.g., from other species, are known and available and can be found in public databases such as Entrez (the National Library of Medicine, Bethesda MD) and EBI- EMBL (Hinxton, Cambridge UK). Examples of such sequences from the UNIPROT database (available at UniProt.org and see The UniProt Consortium, Nucleic Acids Res. D142-D148 (2010)), include the following:
Figure imgf000033_0001
Cytokine domains described herein can also include substitutions present in variant cytokine domains that are able to bind to IL-1 Rl. For example, position 15 of SEQ ID NO:1 (corresponding to position 20 of SEQ ID NO:3) can be Met or Asn.
Position 30 of SEQ ID NO:1 (corresponding to position 34 of SEQ ID NO:3) can be Gly, His, Trp, or Met.
Additional exemplary variants of IL-1 β and IL-1 Ra include those described in Boraschi et al. (1996) Frontiers in Bioscience: A Journal and Virtual Library 1 , d270-308, Evans et al., J. Biol. Chem., 270:1 1477 (1995) and Greenfeder et al., J. Biol. Chem., 270:22460 (1995). For example, variants of IL-1 β include R1 1 G, R1 1 A, Q15H, E105G, and T147G. See, e.g., Evans et al. Variants of IL-1 Ra include W16Y, Q20M, Q20N, Y34G, Y34H, Y34W, Y34M, Y147G, Y147H, Y147M, K145D, H54P, V18S, T108K, C1 16F, C122S, C122A, Y147G, H54P, H54I, and others in Evans et al. and Greenfeder et al., J. Biol. Chem., 270:22460 (1995). A cytokine domain can include a residue identical to IL-1 Ra at one of the foregoing positions. A cytokine domain can also include a residue differing from one of the foregoing mutations at a corresponding position.
In addition, IL-1 family cytokines can include one or more unpaired cysteine residues. One or more, e.g., two, three or all such unpaired cysteine residues can be mutated to another amino acid, e.g., an uncharged amino acid such as alanine or serine. For example, P01 includes cysteines at positions 67, 70, 1 16, and 122 of SEQ ID NO:17. One, two, three or all four such cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine. P02 includes cysteines at positions 67, 70, 1 16, and 122 of SEQ ID NO:18. One, two, three, or all four such cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine. P03 includes cysteines at positions 70, 1 16, and 122 of SEQ ID NO:19. One, two, or all three such cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine. P04 includes cysteines at positions 70, 1 16, and 122 of SEQ ID NO:20. One, two, or all three such cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine. P05 includes cysteines at positions 8, 70, and 122 of SEQ ID NO:21 . One, two, or all three such cysteines can be substituted with another amino acid, e.g., an uncharged amino acid such as alanine or serine.
An IL-1 family cytokine domain, including the chimeric cytokine domains described herein, can also be cyclically permutated. For example, a C-terminal segment from the domain can be repositioned so that it is N-terminal to the original N-terminus and an N-terminal segment (generally comprising the remainder of the original protein after excision of the C-terminal segment). The two repositioned segments can be separated by a linker (e.g., of between about three to ten amino acids). Generally, all the amino acids in the domain prior to permutation are retained except for the change in order. In some embodiments, the cut point for the permutation can be in a flexible region, e.g., a flexible loop such as such as the β6-β7 (e.g., amino acids corresponding to 71 -80 of SEQ ID NO:3) or the β7-β8 loop (e.g., amino acids corresponding to 84-99 of SEQ ID NO:3).
In some embodiments, a receptor binding agent described herein, e.g., a receptor binding agent which includes an IL-1 family cytokine domain has a molecular weight less than 30, 25, 22, 20, 19, 18, or about 17 kDa. In some embodiments, the receptor binding agent has a molecular weight greater than 18, 19, 20, 22, 25, 30, 40, 45, or 50 kDa. For example, the receptor binding agent can include other polypeptide, polymeric or non-polymeric components, for example, components which modify the agents pharmacokinetics, stability, immunogenicity, and/or molecular weight. The protein may include other modifications, e.g., post-translational or synthetic
modifications. In certain embodiments, the receptor binding agent is not glycosylated. In other embodiments, the receptor binding agent includes at least one glycosylation.
For example, receptor binding agents described herein can include additional domains and features. For example, a receptor binding agent can be fused, directly or indirectly, to a domain of an antibody protein, e.g., to an Fc domain or to one or more constant domains (e.g., CH1 , CH2, or CH3). For example, the domains can be human domains or variants of human domains. The Fc domain or the one or more constant domains can be located N-terminal or C-terminal to the receptor binding agent.
Fc domains can be obtained from any suitable immunoglobulin, e.g., from a human antibody, e.g., such as an antibody of the lgG1 , lgG2, lgG3, or lgG4 subtypes, IgA, IgE, IgD or IgM. In one example, the Fc domain includes a sequence from an amino acid residue at about position Cys226, or from about position Pro230, to the carboxyl terminus of the Fc domain. An Fc domain generally includes two constant domains, a CH2 domain and a CH3 domain, and optionally includes a CH4 domain.
Antibodies with substitutions in an Fc region thereof and increased serum half- lives are also described in WO00/42072, WO 02/060919; Shields et al., J. Biol. Chem. 276:6591 - 6604 (2001 ); Hinton, J. Biol. Chem. 279:6213-6216 (2004)). Numbering of the residues in an IgG heavy chain is that of the EU index as in Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, NH1 , MD (1991 ) with reference to the numbering of the human lgG1 EU antibody therein.
In one embodiment, a receptor binding agent includes a salvage receptor-binding epitope which increases in vivo serum half-life, as described, e.g., in US Patent
5,739,277 and Ghetie et al., Ann. Rev. Immunol. 18:739-766 (2000)). In some embodiments, the epitope is include in an Fc region that is fused to the receptor binding agent.
In one embodiment, a receptor binding agent includes a serum albumin sequence, or a portion of such sequence that binds to the FcRn receptor, or a sequence which binds to serum albumin, e.g., human serum albumin. For example, certain peptides bind to serum albumin can be associated with the receptor binding agent, e.g., the sequence DICLPRWGCLW (SEQ ID NO:22). See also, Dennis et al. J. Biol. Chem. 277:35035-35043 (2002).
Receptor binding agents can be modified to include a sequence that increases the size and stability of the agent, e.g., a sequence described in WO2008/155134 or WO2009/023270. Such sequences can be generally biologically inactive, e.g., it does not modulate signaling mediated by IL-1 cytokine family members. A variety of stabilizing polypeptide sequences can be used, e.g., sequences rich in glycine and/or serine, as well as other amino acids such as glutamate, aspartate, alanine or proline. For example, sequences can be designed to have at least 30, 40, 50, 60, 70, 80, 90 or 100% glycine and/or serine residues. In some embodiments, the combined length of stabilizing polypeptide sequences that are attached to a protein can be at least 20, 25, 35, 50, 60, 70, 80, 90, 100, 120, 140, 160, 180, 200, 250, 300, 350, 400, 500, 600, 700, 800, 900 or more than 1000 or 2000 amino acids. Stabilizing sequences can be, for example, fused to a biologically active polypeptide, for example to the N- or C-terminus of the receptor binding agent. Fusion of stabilizing sequences can result in a significant increase in the hydrodynamic radius of the fusion protein relative to the unmodified protein, which can be detected by ultracentrifugation, size exclusion chromatography, or light scattering, for example. In some embodiments, stabilizing sequences to contain few or none of the following amino acids: cysteine (to avoid disulfide formation and oxidation), methionine (to avoid oxidation), asparagine and glutamine (to avoid desamidation) and aspartate. Stabilizing sequences can be designed to contain proline residues that tend to reduce sensitivity to proteolytic degradation. Binding Assays
The interaction of a receptor binding agent and its targets can be analyzed using any suitable approach, including for example radio-immunoassays, cell binding assays, and surface plasmon resonance (SPR). An exemplary cell binding assay using radio- iodinated protein competition is described in Boraschi, J. Immunol., 155(10):4719-25 (1995).
SPR or Biomolecular Interaction Analysis (BIA) can detect biospecific
interactions in real time and without labeling any of the interactants. Changes in the mass at the binding surface (indicative of a binding event) of the BIA chip result in alterations of the refractive index of light near the surface (the optical phenomenon of surface plasmon resonance (SPR)). The changes in the refractivity generate a detectable signal, which are measured as an indication of real-time reactions between biological molecules. Methods for using SPR are described, for example, in Raether, 1988, Surface Plasmons Springer Verlag; Sjolander and Urbaniczky, 1991 , Anal. Chem. 63:2338-2345; Szabo et al., 1995, Curr. Opin. Struct. Biol. 5:699-705 and on-line resources provide by BIAcore International AB (Uppsala, Sweden). A BIACORE® system or a Reichert SR7000DC Dual Channel SPR can be used to compare and rank interactions in real time, in terms of kinetics, affinity or specificity without the use of labels. Binding affinities of a receptor binding agent for a cytokine receptor extracellular domain (e.g., the extracellular domain of IL-1 Rl) can be measured using SPR under approximately physiological conditions, e.g., 10 mM HEPES pH 7.4, 150 mM NaCI, 3 mM EDTA, 0.005% Tween-20. Other methods that do not rely on SPR can also be used, e.g., to measure binding and affinity.
Information from binding assays can be used to provide an accurate and quanti- tative measure of the equilibrium dissociation constant (KD), and kinetic parameters
(e.g., Kon and Koff) for the binding of a receptor binding agent to a target. Such data can be used to compare different proteins, targets, and conditions. This information can also be used to develop structure-activity relationships (SAR). For example, the kinetic and equilibrium binding parameters of variant proteins can be compared to the parameters of a reference or parent protein. Variant amino acids at given positions can be identified that correlate with particular binding parameters, e.g., high affinity and slow Koff. This information can be combined with structural modeling (e.g., using homology modeling, energy minimization, or structure determination by x-ray crystallography or NMR). Proteins used for evaluating affinities can be produced in recombinant form and can include tags suitable for purification or immobilization, e.g., a FLAG tag, myc tag, hemagglutinin tag, His tag, or Fc domain fusion. Extracellular domains of receptor proteins (such as IL-1 Rl, and IL-1 RAcP) can be produced in recombinant form by expression, e.g., in bacterial or insect cells, for example, using baculovirus expression in Sf9 cells. Soluble receptor proteins can be immobilized in the BIAcore system, e.g., using chips that include reagents that bind to their tags, for example, a chip coated with IgG specific for the Fc domain or other tag.
Cellular Activity Assays
The ability of receptor binding agents to function as receptor antagonists can be evaluated, e.g., in a cell based assay. For example, it is possible to evaluate IL-1 Rl inhibition by a receptor binding agent. Several exemplary assays for IL-1 activity are described in Boraschi et al. and include T cell proliferation assays, IL-6 and IL-8 production assays, and inhibition of calcium influx.
In one exemplary assay, the ability of a receptor binding agent is evaluated for its ability to inhibit I L-1 β stimulated release of IL-6 from human fibroblasts. Inhibition of IL- Ι β-stimulated cytokine release in MRC5 cells is correlated with the agent's ability to inhibit IL-1 mediated activity in vivo. Details of the assay are described in Dinarello et al., Current Protocols in Immunology, Ch. 6.2.1 -6.2.7, John Wiley and Sons Inc., 2000. Briefly, human MRC5 human fibroblasts (ATCC # CCL-171 , Manassas VA, USA) are grown to confluency in multi-well plates. Cells are treated with titrated doses of the receptor binding agent and controls. Cells are subsequently contacted with 100 pg/ml of IL-1 β in the presence of the titrated agent and/or controls. Negative control cells are not stimulated with IL-1 β . The amounts of IL-6 released in each group of treated cells is measured using an IL-6 ELISA kit (e.g., BD Pharmingen, Franklin Lakes, NJ, USA). Controls that can be used include buffer alone, IL-1 Ra, and antibodies to IL-1 β.
Efficacy of a receptor binding agent can also be evaluated in vivo. An exemplary assay is described in Economides et al., Nature Med., 9:47-52 (2003). Briefly, mice are injected intraperitoneal^ with titrated doses of the receptor binding agent and controls. Twenty-four hours after injection, mice are injected subcutaneously with recombinant human IL-1 β at a dose of 1 μg kg. Two hours after injection of the IL-1 β (peak IL-6 response time), mice are sacrificed, and blood is collected and processed for serum. Serum IL-6 levels are assayed by ELISA. Percent inhibition can be calculated based on the ratio of IL-6 detected in experimental animal serum to IL-6 detected in controls.
Other exemplary assays for IL-1 activity in vivo are described in Boraschi et al. and include an anorexia, hypoglycemia, and neutrophilia assay. Production
Receptor binding agents can be produced by expression in recombinant host cells, but also by other methods such as in vitro transcription and translation and chemical synthesis.
For cellular expression, one or more nucleic acids (e.g., cDNA or genomic DNA) encoding a receptor binding agent may be inserted into a replicable vector for cloning or for expression. Various vectors are publicly available. The vector may, for example, be a plasmid, cosmid, viral genome, phagemid, phage genome, or other autonomously replicating sequence. The appropriate coding nucleic acid sequence may be inserted into the vector by a variety of procedures. For example, appropriate restriction endonuclease sites can be engineered (e.g., using PCR). Then restriction digestion and ligation can be used to insert the coding nucleic acid sequence at an appropriate location. Vector components generally include one or more of an origin of replication, one or more marker genes, an enhancer element, a promoter, and a transcription termination sequence.
The receptor binding agent may be produced recombinantly either in isolation but also by fusion to one or more other components, such as a signal sequence, an epitope or purification moiety, and a label. The receptor binding agent can include the pro domain of an interleukin-1 family member, e.g., which subsequently can be removed by proteolytic processing.
For bacterial expression, the receptor binding agent can be produced with or without a signal sequence. For example, it can be produced within cells so that it accumulates in inclusion bodies, or in the soluble fraction. It can also be secreted, e.g., by addition of a prokaryotic signal sequence, e.g., an appropriate leader sequence such as from alkaline phosphatase, penicillinase, or heat-stable enterotoxin II. Exemplary bacterial host cells for expression include any transformable £ coli K-12 strain (such as £ coli BL21 , C600, ATCC 23724; £ coli HB101 NRRLB-1 1371 , ATCC-33694; £ coli MM294 ATCC-33625; £ coli W31 10 ATCC-27325), strains of B. subtilis, Pseudomonas, and other bacilli. Proteins produced in bacterial systems will typically lack glycosylation. Accordingly, in some embodiments, the receptor binding agents described herein are substantially free of glycosylation, e.g., free of glycosylation modifications of a mammalian or other eukaryotic cell.
The receptor binding agent can be expressed in a yeast host cell, e.g.,
Saccharomyces cerevisiae, Schizosaccharomyces pombe, Hanseula, or Pichia pastoris. For yeast expression, the receptor binding agent can also be produced intracellular^ or by secretion, e.g., using the yeast invertase leader or alpha factor leader (including Saccharomyces and Kluyveromyces forms), or the acid phosphatase leader, or the C. albicans glucoamylase leader (EP 362,179 published 4 Apr. 1990). In mammalian cell expression, mammalian signal sequences may be used to direct secretion of the protein, such as signal sequences from secreted polypeptides of the same or related species, as well as viral secretory leaders. Alternatively, the receptor binding agent can be produced with a pro domain of an interleukin-1 family member, e.g., an IL-1 a or IL-1 β pro domain.
Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Such sequences are well known for a variety of bacteria, yeast, and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria; the 2μ plasmid origin is suitable for yeast; and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells.
Expression and cloning vectors typically contain a selection gene or marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies (such as the URA3 marker in Saccharomyces), or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli. Various markers are also available for mammalian cells, e.g., DHFR or thymidine kinase. DHFR can be used in conjunction with a cell line (such as a CHO cell line) deficient in DHFR activity, prepared and propagated as described by Urlaub et al., Proc. Natl. Acad. Sci. USA, 77:4216 (1980).
Expression and cloning vectors usually contain a promoter operably linked to the nucleic acid sequence encoding the receptor binding agent to direct mRNA synthesis. Exemplary promoters suitable for use with prokaryotic hosts include the β-lactamase and lactose promoter systems (Chang et al., Nature, 275:615 (1978); Goeddel et al., Nature, 281 :544 (1979)), alkaline phosphatase, a tryptophan (trp) promoter system (Goeddel, Nucleic Acids Res., 8:4057 (1980); EP 36,776), and hybrid promoters such as the tac promoter (deBoer et al., Proc. Natl. Acad. Sci. USA, 80:21 -25 (1983)). Promoters for use in bacterial systems can also contain an appropriately located Shine-Dalgarno sequence. The T7 polymerase system can also be used to drive expression of a nucleic acid coding sequence placed under control of the T7 promoter. See, e.g., the pET vectors (EMD Chemicals, Gibbstown NJ, USA) and host cells, e.g., as described in Novagen User Protocol TB053 available from EMD Chemicals and US 5,693,489. For example, such vectors can be used in combination with BL21 (DE3) cells and BL21 (DE3) pLysS cells to produce protein, e.g., at least 0.05, 0.1 , or 0.3 mg per ml of cell culture. Other cells lines that can be used include DE3 lysogens of B834, BLR, HMS174, NovaBlue, including cells bearing a pLysS plasmid.
Exemplary promoters for use with yeast cells include the promoters for 3- phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem., 255:2073 (1980)) or other glycolytic enzymes (Hess et al., J. Adv. Enzyme Reg., 7:149 (1968); Holland,
Biochemistry, 17:4900 (1978)), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6- phosphate isomerase, and pyruvate kinase. Other exemplary yeast promoters are inducible and have the additional advantage of transcription controlled by growth conditions. Examples of inducible promoters include the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, metallothionein, and enzymes responsible for maltose and galactose utilization.
Expression of mRNA encoding a receptor binding agent from vectors in mammalian host cells can controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, and from heat-shock promoters.
Heterologous promoter systems can also be used, e.g., promoters responsive to tetracycline. See Urlinger, S., et al. (2000) Proc. Natl. Acad. Sci. USA 97(14):7963- 7968. Transcription can also be driven by an enhancer sequence, located in cis or trans. Exemplary mammalian enhancer sequences include those for globin, elastase, albumin, oc-fetoprotein, and insulin. Additional examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers. The enhancer may be spliced into the vector at a position 5' or 3' to the coding sequence for the receptor binding agent, but is preferably located at a site 5' from the promoter.
Expression vectors used in eukaryotic host cells (yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms) can also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5' and, occasionally 3', untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the receptor binding agent. The expression vector may also include one or more intronic sequences.
The receptor binding agent can also be expressed in insect cells, e.g., Sf9 or SF21 cells, e.g., using the pFAST-BAC™ system. Additional exemplary baculovirus expression vectors are available from Invitrogen, Life Technologies, Carlsbad CA, USA. The receptor binding agent can also be expressed in mammalian cells. For example, cell lines of mammalian origin also may be employed. Examples of mammalian host cell lines include the COS-7 line of monkey kidney cells (ATCC CRL 1651 ) (Gluzman et al., Cell 23:175, 1981 ), L cells, C127 cells, 3T3 cells (ATCC CCL 163), Chinese hamster ovary (CHO) cells, HeLa cells, and BHK (ATCC CRL 10) cell lines, and the CV1 /EBNA cell line derived from the African green monkey kidney cell line CV1 (ATCC CCL 70) as described by McMahan et al. (EMBO J. 10: 2821 , 1991 ). Established methods for introducing DNA into mammalian cells have been described (Kaufman, R. J., Large Scale Mammalian Cell Culture, 1990, pp. 1569).
Still other methods, vectors, and host cells suitable for adaptation to the synthesis of receptor binding agent in recombinant cells are described in Molecular Cloning: A Laboratory Manual, Third Ed., Sambrook et al. (eds.), Cold Spring Harbor Press, (2001 ) (ISBN: 0879695773).
Once expressed in cells, receptor binding agents can be recovered from culture medium, inclusion bodies, or cell lysates. Cells can be disrupted by various physical or chemical means, such as freeze-thaw cycling, sonication, mechanical disruption, or cell lysing agents (e.g., detergents).
Receptor binding agents can be purified from other cell proteins or polypeptides that can be found in cell lysates or in the cell medium. Various methods of protein purification may be employed and such methods are known in the art and described for example in Deutscher, Methods in Enzymology, 182 (1990); and Scopes, Protein Purification: Principles and Practice, Springer- Verlag, New York (2010) (ISBN:
1441928332). Exemplary of purification procedures include: by fractionation on an ion- exchange column; ethanol precipitation; reverse phase HPLC; chromatography on silica or on a cation-exchange resin such as DEAE; chromatofocusing; SDS-PAGE;
ammonium sulfate precipitation; gel filtration using, for example, Sephadex G-75; protein A Sepharose columns to remove contaminants such as IgG; and affinity columns (e.g., metal chelating columns to bind epitope-tagged forms of the protein and columns with various ligands to bind any purification moiety that is associated with the receptor binding agent). A purification method can include a combination of two different ion- exchange chromatography steps, e.g., cation exchange chromatograph followed by anion exchange chromatography, or vice versa. Receptor binding agents can be eluted from ion exchange resin by a variety of methods include salt and/or pH gradients or steps. In some embodiments, the receptor binding agent includes a purification moiety (such as epitope tags and affinity handles). Such moieties can be used for affinity chromatography and can be optionally removed by proteolytic cleavage.
Ion exchange chromatography may also be used as an ion exchange separation technique. Ion exchange chromatography separates molecules based on differences between the overall charge of the molecules and can be used to separate intact form of receptor binding agents from other forms of such proteins.
Anionic or cationic substituents may be attached to matrices in order to form anionic or cationic supports for chromatography. Anionic exchange substituents include diethylaminoethyl (DEAE), quaternary aminoethyl (QAE) and quaternary amine (Q) groups. Cationic substitutents include carboxymethyl (CM), sulfoethyl (SE), sulfopropyl (SP), phosphate (P) and sulfonate (S). Cellulose ion exchange resins such as DE23, DE32, DE52, CM-23, CM-32 and CM-52 are available from Whatman Ltd. (Maidstone, Kent, U.K). SEPHADEX™ and other cross-linked ion exchangers are also known. For example, DEAE-, QAE-, CM-, and SP-SEPHADEX™ and DEAE-, Q-, CM- and S- SEPHAROSE™ and SEPHAROSE™ Fast Flow are available from Pharmacia AB. DEAE and CM derivatized ethylene glycol-methacrylate copolymer such as
TOYOPEARL DEAE-650S or M and TOYOPEARL CM-650S or M are available from Toso Haas Co. (Philadelphia, PA, USA).
A cation exchange surface is an ion exchange surface with covalently bound negatively charged ligands, and which thus has free cations for exchange with cations in a solution in contact with the surface. Exemplary surfaces include cation exchange resins, such as those wherein the covalently bound groups are carboxylate or sulfonate. Commercially available cation exchange resins include CMC-cellulose, SP-Sephadex™ and Fast S-Sepharose™ (Pharmacia).
An anion exchange surface is an ion exchange surface with covalently bound positively charged groups, such as quaternary amino groups. An exemplary anion exchange surface is an anion exchange resin, such as DEAE cellulose, TMAE, QAE Sephadex™ and Fast Q Sepharose™ (Pharmacia).
An exemplary purification scheme for a receptor binding agent includes lysing £. coli cells in lysis buffer following by depth filtration. The material is then subject to cation exchange chromatography (CEX). The CEX eluate is then flowed over anion exchange media in an anion exchange chromatography (AEX) step. The AEX FT can be subject to a polishing step. Material can then be processed by ultrafiltration/diafiltration, e.g., to concentrate or desalt the material. Ultrafiltration/diafiltration membranes may be selected based on nominal molecular weight cut-off ("NMWCO") so as to retain the protein in the retentate, while allowing low molecular weight materials such as salts to pass into the filtrate. Any buffering solution or sterile water may be used during the final buffer exchange step, e.g., depending on the desired final pH and conductivity of the product.
A receptor binding agent can be stored in a variety of solutions, including water,
PBS, and buffered solutions. Exemplary buffered solutions include sodium acetate pH 4.5, sodium acetate pH 4.7, sodium acetate pH 4.9, sodium acetate pH 5.1 , sodium acetate pH 5.3, sodium acetate pH 5.5, succinate pH 5.2, succinate pH 5.4, succinate pH 5.6, succinate pH 5.8, histidine pH 5.7, histidine pH 6.0, histidine pH 6.3, histidine 6.6, sodium phosphate pH 6.5, sodium phosphate pH 6.7, sodium phosphate 7.0, sodium phosphate pH 7.3, sodium phosphate pH 7.7, imidazole pH 6.5, imidazole pH 6.8, imidazole pH 7.2, Tris pH 7.0, Tris pH 7.5, Tris pH 7.7. Buffering agents can be present, e.g., at a concentration of about 1 -100mM, 5-50mM, 10-50mM, or 5-25 mM. The solution can further include a salt such as NaCI (e.g., 50 mM, 150 mM, or 250 mM, and ranges therebetween), arginine (e.g., at about 1%, 2%, 3%, 4%, 5%, 7.5%, and ranges therebetween), sucrose (e.g., at about 1 %, 2%, 3%, 4%, 5%, 7.5%, 8.5%, 10%, 15%, and ranges therebetween), and/or glycerol (e.g., at about 0.5%, 1 %, 2%, 3%, 4%, 5%, 7.5%, 8.5%, 10%, 15%, and ranges therebetween). The receptor binding agent can be present in the composition in an amount of at least 50 mg, 100 mg, 500 mg, 1 g, 5 g, 10g, or more.
Analytical Methods
Host cell proteins (HCP) refer to proteins in a preparation that differ from a receptor binding agent, and for example are endogenous proteins of the host cell from which the receptor binding agent was prepared, typically £ coli proteins. Preferably, host cell proteins are present at fewer than 10000, 1000, 900, 800, or 700 ppm (parts per million). HCP can be detected, for example, by ELISA or other detection methods. For example, the ELISA can use polyclonal antibodies to HCPs. An exemplary kit is available from Cygnus Technologies (CN# F410; Southport NC USA). Another exemplary kit uses AlphaScreen technology (AlphaLisa® E. coli HCP Kit, Product Number AL261 C/F, Perkin Elmer, Waltham MA USA).
Material containing or potentially containing a receptor binding agent can be evaluated, e.g., using high pressure liquid chromatography. Exemplary analytical techniques include weak cation exchange high performance liquid chromatography (wCEX-HPLC), size exclusion high performance liquid chromatography (SE-HPLC), reverse phase liquid chromatography, ESI-MS, turbospray ionization mass spectrometry, nanospray ionization mass spectrometry, thermospray ionization mass spectrometry, sonic spray ionization mass spectrometry, SELDI-MS and MALDI-MS.
In several embodiments, the N-terminal region of a receptor binding agent includes sequences identical to peptides from the N-terminal region of IL-1 β, for example, the peptide APVRS (SEQ ID NO:58). Recombinant cells (particularly £ coli cells) expressing such proteins can produce intact protein as well as other isoforms, including the des-Ala isoform and an isoform with an additional methionine (e.g., N- terminal to Ala1 ). Receptor binding agents that include a proline at amino acid position 2 (where the N terminal residue is at position 1 ) can be susceptible to cleavage by £. coli proteases, such as aminopeptidase P which has cleavage specificity for X-PRO. This cleavage can remove the N-terminal amino acid. For example, P03, P04, and P05 have a proline at position 2. The intact forms of P03, P04, and P05 are 153 amino acids in length and begin with alanine whereas the des-Ala species are 152 amino acids in length and begin with proline.
Analytical techniques such as those above can be used to distinguish between intact forms and other forms, e.g., a des-Ala species. An analytical exemplary technique is wCEX-HPLC. For example, P05 can be evaluated using a Dionex ProPac® WCX-10 4x250 mm column (Product Number 054993) as described in Example 9 below. WCX- HPLC peaks can be evaluated by C4 reversed-phase (RP)-HPLC on-line with mass spectrometry. Intact P05 has theoretical mass: 1 7700.4 Da and is detectable as
1 7700.4 Da. The des-Ala species is detectable as 17629.4 Da, which is 71 Da less than the mass of the intact P05. The 71 Da reduction in mass corresponds to removal of a single alanine residue.
Pharmaceutical Compositions
A receptor binding agent can be formulated as a pharmaceutical composition. Typically, the composition is sterile and includes one or more of a buffer, a
pharmaceutically acceptable salt, and an excipient or stabilizer. For example, the composition can be an aqueous composition. A receptor binding agent described herein can be formulated according to standard methods for a biologic. See e.g., Gennaro (ed.), Remington: The Science and Practice of Pharmacy, 20th ed., Lippincott, Williams & Wilkins (2000) (ISBN: 0683306472); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Ed.., Lippincott Williams & Wilkins Publishers (1999) (ISBN: 0683305727); Kibbe (ed.), Handbook of Pharmaceutical Excipients, 3rd ed. (2000) (ISBN: 091733096X); Protein formulation and delivery, McNally and Hastedt (eds.), Informa Health Care (ISBN: 0849379490) (2007).
A receptor binding agent for a pharmaceutical composition is typically at least 10,
20, 50, 70, 80, 90, 95, 98, 99, or 99.99% pure and typically free of human proteins. It can be the only protein in the composition or the only active protein in the composition. It can also be combined with one or more other active proteins, e.g., one or more other purified active proteins, e.g., a related or unrelated protein. In some embodiments, the composition can contain the receptor binding agent at a concentration of between about 0.001 - 10%, e.g., 0.001 - 0.1 %, 0.01 - 1 %, or 0.1 % - 1 0%.
Accordingly, also featured herein are purified and isolated forms of the agents described herein. The term "isolated" refers to material that is removed from its original environment (e.g., the cells or materials from which the receptor binding agent is produced). Pharmaceutical compositions can be substantially free of pyrogenic materials, substantially free of nucleic acids, and/or substantially free of cellular enzymes and components, such as polymerases, ribosomal proteins, and chaperone proteins. A pharmaceutical composition can include a pharmaceutically acceptable carrier. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. A "pharmaceutically acceptable salt" refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (see e.g., Berge, S.M., et al. (1977) J. Pharm. Sci. 66:1 -19), e.g., acid addition salts and base addition salts.
In one embodiment, the receptor binding agent is formulated with one or more excipients, such as sodium chloride, and a phosphate buffer (e.g., sodium dibasic phosphate heptahydrate, sodium monobasic phosphate), and polysorbate. It can be provided, for example, in a buffered solution, e.g., at a concentration of about 5-100, 5- 30, 30-50, or 50-100 mg/ml and can be stored at 2-8qC. Pharmaceutical compositions may also be in a variety of other forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, and liposomes. The preferred form can depend on the intended mode of administration and therapeutic application. Compositions for the agents described herein are typically in the form of injectable or infusible solutions, or are for topical or ocular delivery (see below).
Pharmaceutical compositions typically are sterile and stable under the conditions of manufacture and storage. A pharmaceutical composition can also be tested to ensure it meets regulatory and industry standards for administration. The composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration. Sterile injectable solutions can be prepared by incorporating an agent described herein in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating an agent described herein into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of an agent described herein plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable
compositions can be engineered by inclusion of an agent that delays absorption, for example, monostearate salts and gelatin.
For example, a receptor binding agent can be associated with a polymer, e.g., a substantially non-antigenic polymer, such as a polyalkylene oxide or a polyethylene oxide. In some embodiments, the polymer covalently attached to the receptor binding agent, e.g., directly or indirectly. Suitable polymers will vary substantially by weight. Polymers having molecular number average weights ranging from about 200 to about 35,000 Daltons (or about 1 ,000 to about 15,000, and 2,000 to about 12,500) can be used. For example, a receptor binding agent can be conjugated to a water soluble polymer, e.g., a hydrophilic polyvinyl polymer, e.g. polyvinylalcohol or
polyvinylpyrrolidone. A non-limiting list of such polymers include polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols,
polyoxyethylenated polyols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymers is maintained. Additional useful polymers include polyoxyalkylenes such as polyoxyethylene, polyoxypropylene, and block copolymers of polyoxyethylene and polyoxypropylene (Pluronics);
polymethacrylates; carbomers; branched or unbranched polysaccharides that includee the saccharide monomers D-mannose, D- and L-galactose, fucose, fructose, D-xylose, L-arabinose, D-glucuronic acid, sialic acid, D-galacturonic acid, D-mannuronic acid (e.g. polymannuronic acid, or alginic acid), D-glucosamine, D-galactosamine, D- glucose and neuraminic acid including homopolysaccharides and heteropolysaccharides such as lactose, amylopectin, starch, hydroxyethyl starch, amylose, dextrane sulfate, dextran, dextrins, glycogen, or the polysaccharide subunit of acid mucopolysaccharides, e.g. hyaluronic acid; polymers of sugar alcohols such as polysorbitol and polymannitol;
heparin or heparan.
In certain embodiments, the receptor binding agent may be prepared with a carrier that will protect the compound against rapid release. It may be delivered as a controlled release formulation, delivered by an implant or a microencapsulated delivery system. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. See generally e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978. Administration
A receptor binding agent can be administered to a subject, e.g., a human subject, by a variety of methods, such as intravenous administration as a bolus or by continuous infusion over a period of time, by intramuscular, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, intrasynovial, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural injection, intrasternal injection and infusion. Still other modes of administration include topical (e.g., dermal or mucosal) or inhalation (e.g., intranasal or intrapulmonary) routes. For many applications, the route of administration is one of: intravenous injection or infusion, subcutaneous injection, or intramuscular injection.
A receptor binding agent can be administered as a fixed dose or in a mg/kg dose. It can be administered intravenously (IV) or subcutaneously (SC). The receptor binding agent can administered, for example, every day, every other day, every third, fourth or fifth day, every week, every three to five weeks, e.g., every fourth week, or monthly.
A pharmaceutical composition may include a "therapeutically effective amount" of an agent described herein. A therapeutically effective amount of an agent may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual, e.g., amelioration of at least one disorder parameter, or amelioration of at least one symptom of the disorder (and optionally the effect of any additional agents being administered). A therapeutically effective amount is also one in which any toxic or detrimental effects of the composition are outweighed by the therapeutically beneficial effects. A receptor binding agent is typically administered in a therapeutically effective amount.
Pharmaceutical compositions can be administered using medical devices, e.g., implants, infusion pumps, hypodermic needles, and needleless hypodermic injection devices. The device can include, e.g., one or more housings for storing pharmaceutical compositions, and can be configured to deliver unit doses of the receptor binding agent, and optionally a second agent. The doses can be fixed doses, i.e., physically discrete units suited as unitary dosages for the subjects to be treated; each unit can contain a predetermined quantity of receptor binding agent calculated to produce the desired therapeutic effect in association with a pharmaceutical carrier and optionally in association with another agent.
In some embodiments, to treat a disorder described herein, a receptor binding agent can be administered to the subject having the disorder in an amount and for a time sufficient to induce a sustained improvement in at least one indicator that reflects the severity of the disorder. An improvement is considered "sustained" if the subject exhibits the improvement on at least two occasions separated by one to four weeks. The degree of improvement can be determined based on signs or symptoms, and can also employ questionnaires that are administered to the subject, such as quality-of-life
questionnaires.
Various indicators that reflect the extent of the illness may be assessed for determining whether the amount and time of the treatment is sufficient. The baseline value for the chosen indicator or indicators is established by examination of the subject prior to administration of the first dose of the receptor binding agent. Preferably, the baseline examination is done within about 60 days of administering the first dose.
Improvement can be induced by repeatedly administering a dose of the receptor binding agent until the subject manifests an improvement over baseline for the chosen indicator or indicators. In treating chronic conditions, the degree of improvement may be obtained by repeated administration over a period of at least a month or more, e.g., for one, two, or three months or longer, or indefinitely. In treating an acute condition, the agent can be administered for a period of one to six weeks or even a single dose.
Although the extent of illness after treatment may appear improved according to one or more indicators, treatment may be continued indefinitely at the same level or at a reduced dose or frequency. Treatment may also be discontinued, e.g., upon
improvement or disappearance of symptoms. Once treatment has been reduced or discontinued, it may be resumed if symptoms should reappear.
Treatments
A receptor binding agent such as one that binds to IL-1 Rl and that antagonizes IL-1 signaling, can be used to treat an "IL-1 mediated disorder," which includes any disease or medical condition that is (i) caused at least in part by IL-1 agonism, (ii) is associated with elevated levels or activity of an IL-1 signaling component (such as IL-1 a, IL-1 β , or IL-1 R I ) or elevated IL-1 signaling, and/or (iii) is ameliorated by decreasing IL-1 activity. IL-1 mediated disorders include acute and chronic disorders, including autoimmune disorders and inflammatory disorders. IL-1 mediated disorders include systemic and non-systemic disorders. It is well established that IL-1 a and IL-1 β are potent pro-inflammatory cytokines implicated in infectious responses as well as in inflammatory disease, including rheumatoid arthritis. Increased IL-1 production has been observed in patients with several autoimmune disorders, ischemia, and various cancers, therefore implicating IL-1 in these and related diseases.
See also generally Sims and Smith, The IL-1 family: regulators of immunity, Nature Reviews Immunology, doi:10.1038/nri2691 (2010).
The term "treat" refers to the administration of an agent described herein to a subject, e.g., a patient, in an amount, manner, and/or mode effective to improve a condition, symptom, or parameter associated with a disorder, e.g., a disorder described herein, or to prevent progression of a disorder, to either a statistically significant degree or to a degree detectable to one skilled in the art. The treatment can be to intended to cure, heal, alleviate, relieve, alter, remedy, ameliorate, palliate, improve or affect the disorder, the symptoms of the disorder or the predisposition toward the disorder. An effective amount, manner, or mode can vary depending on the subject and may be tailored to the subject. Exemplary subjects include humans, primates, and other non- human mammals. A receptor binding agent can also be given prophylactically to reduce the risk of the occurrence of a disorder or symptom thereof.
The IL-1 mediated disorder can be an autoimmune disorder. Examples of IL-1 mediated autoimmune disorders include: rheumatoid arthritis, ankylosing spondylitis, Behcet's syndrome, inflammatory bowel diseases (including Crohn's disease and ulcerative colitis), asthma, psoriasis, Type I diabetes, and other disorders identified herein. A receptor binding agent described herein can be administered to a subject having or at risk for such IL-1 mediated autoimmune disorders. The IL-1 mediated disorder can be an inflammatory disorder such as described below. A receptor binding agent described herein can be administered to a subject having or at risk for such IL-1 mediated inflammatory disorders.
Exemplary IL-1 mediated disorders include:
Rheumatoid Arthritis and related arthritides. A receptor binding agent can be used to treat a subject having or at risk for rheumatoid arthritis. Rheumatoid arthritis (RA) is a chronic systemic autoimmune inflammatory disease that affects the synovial membrane in joints and which damages articular cartilage. The pathogenesis of RA is T lymphocyte dependent and can include the production of autoantibodies known as rheumatoid factors. Complexes of rheumatoid factor and antigen can form and accumulate in joint fluid and blood, inducing the infiltration of lymphocytes, neutrophils, and monocytes into the synovium. Joints are typically affected in a symmetrical pattern, but extra-articular disease may also occur, e.g., causing pulmonary fibrosis, vasculitis, and cutaneous ulcers, or Felty's syndrome which manifests as neutropenia,
thrombocytopenia and splenomegaly. Patients can also exhibit rheumatoid nodules in the area of affected joints, pericarditis, pleuritis, coronary arteritis, and interstitial pneumonitis with pulmonary fibrosis. A form of IL-1 Ra is indicated for moderately to severely active RA. See, e.g., Cohen, S. et al. Arthritis & Rheumatism 46, 614-24
(2002); Fleischmann, R.M. et al. Arthritis & Rheumatism 48, 927-34 (2003); Nuki, G., et al. Arthritis & Rheumatism 46, 2838-46 (2002); Schiff, M.H. et al. Arthritis &
Rheumatism 50, 1752-60 (2004).
Symptoms of active RA include fatigue, lack of appetite, low grade fever, muscle and joint aches, and stiffness. Muscle and joint stiffness are usually most notable in the morning and after periods of inactivity. During flares, joints frequently become red, swollen, painful, and tender, generally as a consequence of synovitis. Scales useful for assessing RA and symptoms of RA include the Rheumatoid Arthritis Severity Scale (RASS; Bardwell et al., (2002) Rheumatology 41 (1 ):38-45), SF-36 Arthritis Specific Health Index (ASHI; Ware et al., (1999) Med. Care. 37(5 Suppl):MS40-50), Arthritis
Impact Measurement Scales or Arthritis Impact Measurement Scales 2 (AIMS or AIMS2; Meenan et al. (1992) Arthritis Rheum. 35(1 ):1 -10); the Stanford Health Assessment Questionnaire (HAQ), HAQII, or modified HAQ (see, e.g., Pincus et al. (1983) Arthritis Rheum. 26(1 1 ):1346-53).
A receptor binding agent described herein can be administered to a subject having or at risk for RA to delay onset and/or ameliorate one or more of the foregoing signs and symptoms. The subject can have moderate to severe active RA. The subject can be a non-responder to TNF inhibitor therapy (e.g., therapy with ENBREL®
(etanercept), HUMIRA® (adalimumab), or REMICADE® (infliximab)); the subject can have previously been administered a TNF inhibitor; or the subject can also be continuing to receive a TNF inhibitor (and responding or not responding).
The subject can also be administered methotrexate. The subject can be administered one or more other DMARDS (disease modifying anti-rheumatic drugs), a corticosteroid, and/or a non-steroidal anti-inflammatory. Still other drugs which can be co-administered with a receptor binding agent include inhibitors of CD28 (e.g., CTLA4- Ig), inhibitors of RANKL, IFNy, IL-6, IL-8, and IL-17. Inhibitors include antibodies to such mediators, soluble receptors specific for such mediators, and/or antibodies to receptors for such mediators. Juvenile Chronic Arthritis. A receptor binding agent can be used to treat juvenile chronic arthritis, e.g., in a subject that is less than 21 , 18, 17, 16, 15, or 14 years of age. Juvenile chronic arthritis resembles RA in several respects. Subjects can be rheumatoid factor positive. Subjects may have pauciarticular, polyarticular, or systemic forms of the disease. The arthritis can cause joint ankylosis and retarded growth and can also lead to chronic anterior uveitis and systemic amyloidosis. A receptor binding agent can be used delay onset of or ameliorate one or more such symptoms.
A receptor binding agent can be used to treat juvenile idiopathic arthritis, including systemic onset juvenile idiopathic arthritis (SO-JIA). Subjects may have failed prior corticosteroid treatment or required corticosteroid treatment at a daily dose equal to or over 0.3 mg/kg. See e.g., Quartier et al. (201 1 ) Ann Rheum Dis. 70(5)747-54.
Other Rheumatic Disorders. A receptor binding agent described herein can also be used to treat other rheumatic disorders including scleroderma, systemic lupus erythematosus, gout, osteoarthritis, polymyalgia rheumatica, psoriatic arthritis and chronic Lyme arthritis, inflammation of the voluntary muscle and other muscles, including dermatomyositis, inclusion body myositis, polymyositis, and lymphangioleimyomatosis, pyrogenic arthritis syndrome, pediatric granulomatous arthritis (PGA)/Blau syndrome, and other rheumatic disorders discussed herein.
A receptor binding agent can be used to treat metabolic rheumatic disorders, e.g., disorders associated with hyperuricemia, e.g., gout including chronic acute gout, and other crystal-mediated arthropathies. The agent can be used to treat drug-induced flares associated with gout, including for example flares induced by xanthine oxidase inhibitors, urate oxidase, or uricosuric agents. In gout, crystals of uric acid can activate the inflammasome and trigger the release of I L- 1 β .
Spondyloarthropathies. A receptor binding agent can be used to treat spondyloarthropathies, which include disorders such as ankylosing spondylitis, Reiter's syndrome, arthritis associated with inflammatory bowel disease, spondylitis associated with psoriasis, juvenile onset spondyloarthropathy and undifferentiated
spondyloarthropathy. Spondyloarthropathies are frequently associated with the HLA- B27 gene. Subjects may lack rheumatoid factor and may exhibit sacroileitis with or without spondylitis and inflammatory asymmetric arthritis. Subjects may also have ocular inflammation (see below).
Scleroderma. A receptor binding agent can be used to treat scleroderma or systemic sclerosis. Scleroderma is characterized by induration of the skin which may be localized or systemic. Vascular lesions and endothelial cell injury in the microvascul- ature can also be present. Subjects may exhibit mononuclear cell infiltrates in the cutaneous lesions and have anti-nuclear antibodies. Other organs that show pathogenesis can include: the gastrointestinal tract which may have smooth muscle atrophy and fibrosis resulting in abnormal peristalsis/motility; the kidney which may have concentric subendothelial intimal proliferation affecting small arcuate and interlobular arteries with resultant reduced renal cortical blood flow, and which can cause
proteinuria, azotemia and hypertension; skeletal muscle which may involve atrophy, interstitial fibrosis, inflammation, lung, interstitial pneumonitis and interstitial fibrosis; and heart which can exhibit, e.g., contraction band necrosis and scarring/fibrosis. A receptor binding agent described herein can be administered to a subject having or at risk for scleroderma to ameliorate one or more of the foregoing signs and symptoms.
Sjogren's Syndrome. A receptor binding agent can be used to treat Sjogren's syndrome. Sjogren's syndrome is characterized by immune-mediated inflammation and subsequent functional destruction of the tear glands and salivary glands. The disease can be associated with or accompanied by inflammatory connective tissue diseases. The disease is associated with autoantibody production against Ro and La antigens, both of which are small RNA-protein complexes. Lesions can result in
keratoconjunctivitis sicca, xerostomia, with other manifestations or associations including biliary cirrhosis, peripheral or sensory neuropathy, and palpable purpura. Treatment of ocular disorders associated with Sjogren's is also discussed below.
Thyroid Disorders. A receptor binding agent can be used to treat a thyroid disorder. Exemplary thyroid disorders include Graves' disease, Hashimoto's thyroiditis, juvenile lymphocytic thyroiditis, and atrophic thyroiditis, and are the result of an autoimmune response against thyroid antigens with production of antibodies that react with proteins present in and often specific for the thyroid gland. Experimental models are available including spontaneous models: rats (BUF and BB rats) and chickens (obese chicken strain) and inducible models created by immunization of animals with either thyroglobulin, thyroid microsomal antigen (thyroid peroxidase).
Diabetic & Metabolic disorders. A receptor binding agent can be used to treat a diabetic disorder such as juvenile onset diabetes (including autoimmune diabetes mellitus and insulin-dependent types of diabetes) and maturity onset diabetes (including non-insulin dependent and obesity-mediated diabetes), type I diabetes and type II diabetes. For example, type I diabetes mellitus or insulin-dependent diabetes is associated with the autoimmune destruction of pancreatic islet cells caused by autoantibodies and auto-reactive T cells. In addition, reducing IL-1 β activity can improve glucose control and beta-cell function and can be used to treat type II diabetes. See e.g. Owyang et al. Endocrinology. 2010;151 (6):2515-27. For example, in some
embodiments, a receptor binding agent described herein can be administered to a subject that is not being administered insulin, e.g., the subject is not insulin dependent. For example, the subject can be pre-diabetic. The subject can have either impaired glucose tolerance or impaired fasting glucose. The subject can be obese or have a body mass index that is above 23, 25, 30, 35, or 40 kg/m2. The subject can be insulin resistant, and/or characterized by hyperglycemia or hyperinsulinemia. The subject can be at risk for progression to type II diabetes. See also Larsen, et al. (2007) NEJM 356:1517-26 and Larsen, et al. (2009). Diabetes Care 32:1663-8. In some
embodiments, the subject has a fasting plasma glucose of greater than 6.1 , 6.5, 7, or 8 mmol/L. In some embodiments, the subject has an A1 C level greater than 5.5, 5.7, 6, 6.4, 7, 7.5 or 8%.
In some embodiments, the subject is also administered an insulin secretagogue, e.g., such as an sulfonylurea (e.g., chlorpropamide, tolazamide, acetohexamide, tolbutamide, glyburide, glimepiride, glipizide) and/or meglitinides (e.g., repaglinide, nateglinide) that stimulate insulin secretion. The subject can also be administered a biguanide (e.g., metformin). The receptor binding agent can be administered to reduce loss of and/or damage to pancreatic beta cells.
In some embodiments, the subject is heterozygous or homozygous for the C allele of rs4251961 , located near the 5' of the gene IL1 RN.
Treatment with a receptor binding agent includes ameliorating or preventing deterioration of secondary conditions associated with diabetes, such as diabetic retinopathy, kidney transplant rejection in diabetic patients, obesity-mediated insulin resistance, and renal failure, which itself may be associated with proteinuria and hypertension.
Gastrointestinal Disorders. Receptor binding agents described herein can be used to treat a inflammatory gastrointestinal disorder, including for example coeliac disease, Crohn's disease, ulcerative colitis, idiopathic gastroparesis pancreatitis including chronic pancreatitis, acute pancreatitis, inflammatory bowel disease and ulcers including gastric and duodenal ulcers. Pulmonary disorders. A receptor binding agent can be used to treat a pulmonary disease mediated by IL-1 . Exemplary pulmonary diseases that can be treated include chronic obstructive pulmonary disease (e.g. emphysema and chronic bronchitis), pulmonary alveolar proteinosis, bleomycin-induced pneumopathy and fibrosis, pulmonary fibrosis, including idiopathic pulmonary fibrosis and radiation-induced pulmonary fibrosis, pulmonary sarcoidosis, cystic fibrosis, collagen accumulation in the lungs, ARDS, broncho-pulmonary dysplasia (BPD), chronic obstructive pulmonary diseases, and chronic fibrotic lung disease of preterm infants. In addition, the receptor binding agents described herein can be used to treat occupational lung diseases, including asbestosis, coal worker's pneumoconiosis, silicosis or similar conditions associated with long-term exposure to fine particles. Inflammatory and fibrotic lung disease including eosinophilic pneumonia, idiopathic pulmonary fibrosis, and
hypersensitivity pneumonitis may involve a misregulated immune-inflammatory response that can be treated using a receptor binding agent.
Sarcoidosis is a condition of unknown etiology which is characterized by the presence of epithelioid granulomas in nearly any tissue in the body; involvement of the lung is most common. The pathogenesis involves the persistence of activated macrophages and lymphoid cells at sites of the disease with subsequent chronic sequelae resultant from the release of locally and systemically active products released by these cell types.
Cardiovascular Disorders. A receptor binding agent described herein can be used to treat a cardiovascular disorder or injury, such as aortic aneurysms, acute coronary syndrome, arteritis, vascular occlusion, including cerebral artery occlusion, complications of coronary by-pass surgery, ischemia/reperfusion injury, heart disease, including atherosclerotic heart disease, myocarditis, including chronic autoimmune myocarditis and viral myocarditis, heart failure, including chronic heart failure, congestive heart failure, myocardial infarction, restenosis and/or atherosclerosis after heart surgery or after carotid artery balloon angioplastic procedures, silent myocardial ischemia, left ventricular pump dysfunction, post implantation complications of left ventricular assist devices, Raynaud's phenomena, thrombophlebitis, vasculitis including Kawasaki's vasculitis, veno-occlusive disease, giant cell arteritis, Wegener's granulomatosis, and Schoenlein-Henoch purpura. The receptor binding agent can also be provided prophylactically, e.g., to reduce risk of such cardiovascular disorder. In some embodiments, the receptor binding agent is administered to a patient to treat
atherosclerosis or to reduce risk thereof.
IL-1 mediated signaling is activated by acute myocardial infarction and can initiate apoptotic cell death in the peri-infarct myocardial cells, extending the size of the infarct zone. A receptor binding agent described herein can be administered to reduce damage caused by myocardial infarction. For example, the receptor binding agent can be administered a subject who is at risk for a myocardial infarction, or a subject who has experienced a myocardial infarction, particularly an acute myocardial infarction, e.g., within the last 2, 4, 6, 12, 24, or 48 hours. The agent can be administered in
combination with other agents, including, e.g., heparin and aspirin.
A receptor binding agent can also be used to treat stroke, sub-arachnoid hemorrhage, head trauma or brain injury, and/or inflammation associated with a cardiovascular disorder. For example, elevated levels of IL-1 β have been implicated neuroinflammation associated with stroke and brain injury (Rothwell, N. J., et al., TINS 23(12): 618-625, 2000). The receptor binding agent can be administered to reduce such inflammation and other inflammation associated with ischemia and/or hypoxia. In addition, the receptor binding agent can also be provided prophylactically, e.g., to reduce risk of such disorders and/or inflammation associated with such disorders. For example, the receptor binding agent can be administered a subject who is at risk for a stroke, an ischemic event, other cardiovascular event, or a hemorrhagic event (such as a subarachnoid hemorrhage), or a subject who has experienced a stroke, an ischemic event, other cardiovascular event, or a hemorrhagic event (such as a sub-arachnoid
hemorrhage), e.g., within the last 2, 4, 6, 12, 24, or 48 hours.
Genitourinary and Renal Disorders. Disorders of the genitourinary system can also be treated with a receptor binding agent described herein. Such disorders include glomerulonephritis, including autoimmune glomerulonephritis, glomerulonephritis due to exposure to toxins or glomerulonephritis secondary to infections with haemolytic streptococci or other infectious agents. Immune mediated renal diseases, including glomerulonephritis and tubulointerstitial nephritis, are the result of antibody or T lymphocyte mediated injury to renal tissue either directly as a result of the production of autoreactive antibodies or T cells against renal antigens or indirectly as a result of the deposition of antibodies and/or immune complexes in the kidney that are reactive against other, non-renal antigens. Thus other immune-mediated diseases that result in the formation of immune-complexes can also induce immune mediated renal disease as an indirect sequelae. Both direct and indirect immune mechanisms result in
inflammatory response that produces/induces lesion development in renal tissues with resultant organ function impairment and in some cases progression to renal failure. Both humoral and cellular immune mechanisms can be involved in the pathogenesis of lesions.
A receptor binding agent can also be used to treat uremic syndrome and its clinical complications (for example, renal failure, anemia, and hypertrophic
cardiomyopathy), including uremic syndrome associated with exposure to environmental toxins, drugs or other causes. Complications that arise from inflammation of the gallbladder wall that leads to alteration in absorptive function can also be treated.
Included in such complications are cholelithiasis (gallstones) and choliedocholithiasis (bile duct stones) and the recurrence of cholelithiasis and choliedocholithiasis. Further conditions that can be treated are complications of hemodialysis; prostate conditions, including benign prostatic hypertrophy, nonbacterial prostatitis and chronic prostatitis; and complications of hemodialysis. A receptor binding agent can also be used to treat chronic pain conditions, such as chronic pelvic pain, including chronic prostatitis/pelvic pain syndrome, and post-herpetic pain.
Hematologic and Oncologic Disorders. A receptor binding agent described herein can be used to treat various forms of cancer, including acute myelogenous leukemia, chronic myelogenous leukemia, Epstein-Barr virus-positive nasopharyngeal carcinoma, glioma, colon, stomach, prostate, renal cell, cervical and ovarian cancers, lung cancer (SCLC and NSCLC), including cancer-associated cachexia, fatigue, asthenia, paraneoplastic syndrome of cachexia and hypercalcemia. See, e.g., Voronov et al. (2003) PNAS 100:2645-2650. Solid tumors, including sarcoma, osteosarcoma, and carcinoma, such as adenocarcinoma (for example, breast cancer) and squamous cell carcinoma are also treatable. Regarding the role of IL-1 β in certain tumors, see e.g., Krelin et al. (2007) Cancer Res. 67:1062-1071 . Additional cancers include esophogeal cancer, gastric cancer, gall bladder carcinoma, leukemia, including acute myelogenous leukemia, chronic myelogenous leukemia, myeloid leukemia, chronic or acute
lymphoblastic leukemia and hairy cell leukemia. Other malignancies with invasive metastatic potential, including multiple myeloma, can be treated with the receptor binding agents. See, e.g., Lust et al. (2009) Mayo Clin Proc 84(2):1 14-122.
A receptor binding agent can also be used to treat anemias and hematologic disorders, including chronic idiopathic neutropenia, anemia of chronic disease, aplastic anemia, including Fanconi's aplastic anemia; idiopathic thrombocytopenic purpura (ITP); thrombotic thrombocytopenic purpura, myelodysplastic syndromes (including refractory anemia, refractory anemia with ringed sideroblasts, refractory anemia with excess blasts, refractory anemia with excess blasts in transformation); myelofibrosis/myeloid metaplasia; and sickle cell vasocclusive crisis.
Autoimmune hemolytic anemia, immune pancytopenia, and paroxysmal nocturnal hemoglobinuria can result from production of antibodies that react with antigens expressed on the surface of red blood cells (and in some cases other blood cells including platelets as well) and is a consequence of the removal of those antibody coated cells via complement mediated lysis and/or ADCC/Fc-receptor-mediated mechanisms. In autoimmune thrombocytopenia including thrombocytopenic purpura, and immune-mediated thrombocytopenia in other clinical settings, platelet
destruction/removal occurs as a result of either antibody or complement attaching to platelets and subsequent removal by complement lysis, ADCC or FC-receptor mediated mechanisms.
The receptor binding agent can also be administered to subjects having or at risk for various lymphoproliferative disorders, including autoimmune lymphoproliferative syndrome (ALPS), chronic lymphoblastic leukemia, hairy cell leukemia, chronic lymphatic leukemia, peripheral T-cell lymphoma, small lymphocytic lymphoma, mantle cell lymphoma, follicular lymphoma, Burkitt's lymphoma, Epstein-Barr virus-positive T cell lymphoma, histiocytic lymphoma, Hodgkin's disease, diffuse aggressive lymphoma, acute lymphatic leukemias, T gamma lymphoproliferative disease, cutaneous B cell lymphoma, cutaneous T cell lymphoma (i.e., mycosis fungoides) and Sezary syndrome.
Hepatic Disorders. The receptor binding agents disclosed herein are also useful for treating conditions of the liver such as hepatitis, including acute alcoholic hepatitis, acute drug-induced or viral hepatitis, hepatitis A, B and C, sclerosing cholangitis, hepatic sinusoid epithelium, and inflammation of the liver due to unknown causes.
Hearing Disorders. Receptor binding agents can also be used to treat disorders that involve hearing loss and that are associated with abnormal IL-1 expression. Such disorders include cochlear nerve-associated hearing loss that is thought to result from an autoimmune process, e.g., autoimmune hearing loss, Meniere's syndrome and cholesteatoma, a middle ear disorder often associated with hearing loss. Bone Disorders. Non-arthritic disorders of the bones and joints and also treatable with the receptor binding agents described herein. This encompasses inflammatory disorders of the bone or joint, osteoclast disorders that lead to bone loss, such as but not limited to osteoporosis, including post-menopausal osteoporosis, osteoarthritis, periodontitis resulting in tooth loosening or loss, and prosthesis loosening after joint replacement (generally associated with an inflammatory response to wear debris), e.g., orthopedic implant osteolysis.
Amyloid Disorders. Further, the receptor binding agents described herein can be used to treat primary amyloidosis and the secondary amyloidosis that is characteristic of various conditions, including Alzheimer's disease, secondary reactive amyloidosis; Down's syndrome; and dialysis-associated amyloidosis. In addition, receptor binding agents can be used to treat Amyotrophic lateral sclerosis (ALS), Huntington's disease, and Parkinson's disease. These diseases can also involve formation of aggregates and amyloids that can trigger inflammatory responses.
Neurological Disorders. Receptor binding agents can also be used to treat neuroinflammation and demyelinating diseases of the central and peripheral nervous systems, including multiple sclerosis; idiopathic demyelinating polyneuropathy or Guillain-Barre syndrome; and chronic inflammatory demyelinating polyneuropathy. These disorders are believed to have an autoimmune basis and result in nerve demyelination as a result of damage caused to oligodendrocytes or to myelin directly. In multiple sclerosis disease induction and involve T lymphocytes. Multiple sclerosis has either a relapsing-remitting course or a chronic progressive course. Lesions contain infiltrates of predominantly T lymphocyte mediated, microglial cells and infiltrating macrophages; CD4+ T lymphocytes are the predominant cell type at lesions. The mechanism of oligodendrocyte cell death and subsequent demyelination is not known but is likely T lymphocyte driven.
Myopathies. Receptor binding agents can be used to treat myopathies associated with inflammation and autoimmunity. Idiopathic inflammatory myopathies including dermatomyositis, polymyositis and others are disorders of chronic muscle inflammation of unknown etiology resulting in muscle weakness. Muscle
injury/inflammation is often symmetric and progressive. Autoantibodies are associated with most forms. These myositis-specific autoantibodies are directed against and inhibit the function of components, proteins and RNA's, involved in protein synthesis. Vasculitis Disorders. A receptor binding agent described herein can be used to treat a vasculitis disorder, e.g., a systemic vasculitis. Systemic vasculitis includes diseases in which the primary lesion is inflammation and subsequent damage to blood vessels which results in ischemia/necrosis/degeneration to tissues supplied by the affected vessels and eventual end-organ dysfunction in some cases. Vasculitides can also occur as a secondary lesion or sequelae to other immune-inflammatory mediated diseases such as rheumatoid arthritis, systemic sclerosis, etc., particularly in diseases also associated with the formation of immune complexes.
Diseases in the primary systemic vasculitis group include: systemic necrotizing vasculitis: polyarteritis nodosa, allergic angiitis and granulomatosis, polyangiitis;
Wegener's granulomatosis; lymphomatoid granulomatosis; and giant cell arteritis.
Miscellaneous vasculitides include: mucocutaneous lymph node syndrome (MLNS or Kawasaki's disease), isolated CNS vasculitis, Behcet's disease, thromboangiitis obliterans (Buerger's disease) and cutaneous necrotizing venulitis. The pathogenic mechanism of these vasculitis disorders is believed to be primarily due to the deposition of immunoglobulin complexes in the vessel wall and subsequent induction of an inflammatory response either via ADCC, complement activation, or both.
CAPS. A receptor binding agent can be used to treat a CAPS disorder, i.e., CIAS1 Associated Periodic Syndromes. CAPS includes three genetic syndromes:
Neonatal Onset Multisystem Inflammatory Disorder (NOMID), Muckle-Wells Syndrome (MWS), and Familial Cold Autoinflammatory Syndrome (FCAS). (Hoffman et al. 2001 Naure 29:301 -305; Feldmann et al. 2002 Am J Hum Genet 71 :198-203; Aksentijevich et al. 2002 Arthritis Rheum 46:3340-3348). CAPS are inherited in an autosomal dominant manner with a sporadic or familial pattern. CIAS1 encodes NALP3, a protein component of the "inflammasome", a subcellular enzyme complex that regulates the activity of caspase 1 . Mutations in CIAS1 lead to increased production of IL-1 and numerous pathological consequences (Aksentijevich et al. 2002 supra). IL-1 strongly induces the production of acute phase reactants in the liver, such as C-reactive protein (CRP) and serum amyloid A (SAA).
CAPS disorders share common clinical features and present as a spectrum of clinical severity. NOMID is the most seriously disabling, MWS somewhat less so and FCAS is the least severe. CAPS disorders have several overlapping features and individuals can have unique constellations of signs and symptoms. Features common to all these conditions include fevers, urticaria-like rash, arthritis or arthralgia, myalgia, malaise, and conjunctivitis.
In NOMID, chronic aseptic meningitis may lead to mental retardation and these patients may also suffer disfiguring and disabling bony overgrowth at the epiphyses and patellae. These patients may also suffer blindness due to optic nerve atrophy that results from increased intracranial pressure. MWS and NOMID are commonly associated with severe inflammation that may include the auditory system, meninges, and joints. These patients may suffer daily high spiking fevers and a chronic rash that frequently changes in distribution and intensity. Patients may suffer hearing loss or deafness. Conjunctivitis and papilledema are frequently observed. Amyloidosis may develop and lead to renal failure due to chronic inflammation and overproduction of acute phase reactants (particularly SM). A receptor binding agent can be administered to a subject having NOMID, MWS, or FCAS or diagnosed as have a genotype associated with NOMID, MWS, or FCAS. In addition, a receptor binding agent can be administered to a subject having TRAPS (TNF receptor associated periodic syndrome).
Dermatological disorders. A receptor binding agent can be used to treat a dermatological disorder, such as an inflammatory dermatological disorder or an autoimmune or immune-mediated skin disease. An exemplary disorder is psoriasis. Additional autoimmune or immune-mediated skin disease including bullous skin diseases, erythema multiforme, and contact dermatitis are mediated by auto-antibodies, the genesis of which is T lymphocyte-dependent. Psoriasis is a T lymphocyte-mediated inflammatory disease. Lesions contain infiltrates of T lymphocytes, macrophages and antigen processing cells, and some neutrophils.
Additional disorders of the skin or mucous membranes that can be treated include acantholytic diseases, including Darier's disease, keratosis follicularis and pemphigus vulgaris. Further additional disorders include: acne, acne rosacea, alopecia areata, aphthous stomatitis, bullous pemphigoid, burns, eczema, erythema, including erythema multiforme and erythema multiforme bullosum (Stevens-Johnson syndrome), inflammatory skin disease, lichen planus, linear IgA bullous disease (chronic bullous dermatosis of childhood), loss of skin elasticity, mucosal surface ulcers, including gastric ulcers, neutrophilic dermatitis (Sweet's syndrome), dermatomyositis, pityriasis rubra pilaris, psoriasis, pyoderma gangrenosum, multicentric reticulohistiocytosis, and toxic epidermal necrolysis. Other skin related conditions treatable by receptor binding agents include dermatitis herpetiformis (Duhring's disease), atopic dermatitis, contact dermatitis, and urticaria (including chronic idiopathic urticaria).
Allergic Disorders. A receptor binding agent can be used to treat an allergic disorder, such as asthma, allergic rhinitis, atopic dermatitis, food hypersensitivity, allergic conjunctivitis (see also below) and urticaria. These diseases are frequently mediated by T lymphocyte induced inflammation, IgE mediated-inflammation, or both.
Asthma is a chronic condition involving the respiratory system in which the airway occasionally constricts, becomes inflamed, and is lined with excessive amounts of mucus, often in response to one or more triggers. Episodes may be triggered by such things as exposure to an environmental stimulant (or allergen) such as cold air, warm air, moist air, exercise or exertion, emotional stress, and viral illness. Airway narrowing causes symptoms such as wheezing, shortness of breath, chest tightness, and coughing. A receptor binding agent can be used to treat asthma and can be formulated for topical or pulmonary delivery for such treatment or can be delivered parenterally.
Transplantation. A receptor binding agent can be administered to a subject that is about to undergo, is undergoing, or is recovering from a transplantation.
Transplantation associated diseases, including graft rejection and graft-versus-host- disease (GVHD), are T lymphocyte-dependent; inhibition of T lymphocyte function is ameliorative. Corneal transplantation can be associated with neovascularization which can be ameliorated by treatment with a receptor binding agent. A receptor binding agent can be also used to treat complications resulting from solid organ transplantation, such as heart, liver, skin, kidney, lung (lung transplant airway obliteration) or other transplants, including bone marrow transplants.
Infectious Diseases. The receptor binding agents described herein are useful for treating protozoal diseases, including malaria and schistosomiasis and to treat erythema nodosum leprosum; bacterial or viral meningitis; tuberculosis, including pulmonary tuberculosis; and pneumonitis secondary to a bacterial or viral infection including influenza infection and infectious mononucleosis.
Also treatable with a receptor binding agent are inherited periodic fever syndromes, including familial Mediterranean fever, hyperimmunoglobulin D and periodic fever syndrome and TNF-receptor associated periodic syndromes (TRAPS), and Adult- onset Still's disease, Schnitzler's syndrome, and fibrosing alveolitis. In some embodiments, a receptor binding agent is administered to a subject to reduce activity or expression of IL-6 in the subject. For example, the subject can have a disorder that is associated or mediated at least in part by IL-6.
Ocular Disorders and Ocular Delivery
The receptor binding agents described herein can be used to treat ocular disorders, including ocular disorders affecting the surface of the eye, inflammatory ocular disorders, and ocular disorders mediated at least in part by an autoimmune reaction.
In some embodiments, the ocular disorder is a dry eye disorder that affects the surface of the eye. The disorder includes conditions also referred to keratoconjunctivitis sicca, keratitis sicca, sicca syndrome, xerophthalmia, tear film disorder, decreased tear production, aqueous tear deficiency, and Meibomian gland dysfunction. Dry eye can include forms that are associated with Sjogren's syndrome (SS), e.g., Sjogren's syndrome associated keratoconjunctivitis sicca, but also forms that are not so
associated, e.g., non- Sjogren's syndrome associated keratoconjunctivitis sicca. The patient may or may not have other manifestations of a systemic autoimmune disorder. IL-1 has been implicated in the pathogenesis of dry eye disorders. See, e.g., Enriquez de Salamanca et al. (2010), Mol. Vis. 16:862-873.
Subjects having a dry eye syndrome can exhibit inflammation of the eye dry, and can experience scratchy, stingy, itchy, burning or pressured sensations, irritation, pain, and redness. Dry eye can be associated with both excessive eye watering and conversely insufficient tear production. A receptor binding agent can be administered to such subjects to ameliorate or prevent the onset or worsening of one or more such symptoms. A receptor binding agent can also be used to mitigate pain, e.g., ocular pain, such as due to neuroinflammation, in a subject who is experiencing such pain.
In some embodiments, the ocular disorder is an ocular disorder associated with a systemic autoimmune disorder (such as Sjogren's syndrome and rheumatoid arthritis) or with a disorder associated with IL-1 or another IL-1 cytokine family member. The patient may or may not have a systemic autoimmune disorder or other manifestations of a systemic autoimmune disorder.
A receptor binding agent can also be used to treat other disorders affecting the surface of the eye, such as the cornea. Such disorders include corneal ocular surface inflammatory conditions, corneal neovascularization, keratitis, including peripheral ulcerative keratitis and microbial keratitis. The receptor binding agent can be used to treat a subject undergoing corneal wound healing (e.g., a subject having a corneal wound). A receptor binding agent can be administered to a subject who is about to receive, undergoing, or recovering from a procedure involving the eye, e.g., corneal transplantation/ keratoplasty, keratoprosthesis surgery, lamellar transplantation, selective endothelial transplantation. See, e.g., Dana (2007) Trans Am Ophthalmol Soc 105: 330-43; Dekaris et al. (1999) Curr Eye Res 19(5): 456-9; and Dana et al. (1997) Transplantation 63:1501 -7. A receptor binding agent can be used to treat disorders affecting the conjunctiva, including conjunctival scarring disorders and conjunctivitis. The receptor binding agent can be used to treat still other disorders such as pemphigoid syndrome and Stevens-Johnson syndrome. A receptor binding agent described herein can be administered to a subject to modulate neovascularization in or around the eye. See, e.g., Dana (2007) Trans Am Ophthalmol Soc 105: 330-43.
A receptor binding agent can be administered to a subject having an allergic reaction affecting the eye, e.g., a subject experiencing severe allergic (atopic) eye disease such as allergic conjunctivitis. For example, the receptor binding agent can be administered topically. See also, e.g., Keane-Myers AM et al. (1999) Invest Ophthalmol Vis Sci, 40(12): 3041 -6.
A receptor binding agent can be administered to a subject having an autoimmune disorder affecting the eye. Exemplary autoimmune ocular disorders include sympathetic ophthalmia, Vogt-Koyanagi Harada (VKH) syndrome, birdshot retinochoriodopathy, ocular cicatricial pemphigoid, Fuchs' heterochronic iridocyclitis, and various forms of uveitis. A receptor binding agent can be administered to a subject to treat any of the foregoing disorders.
A receptor binding agent can be administered to a subject who has or is at risk for diabetic retinopathy. See, e.g., Demircan et al. (2006) Eye 20:1366-1369 and Doganay et al. (2006) Eye, 16:163-170
Uveitis includes acute and chronic forms and includes inflammation of one or more of the iris, the ciliary body, and the choroid. Chronic forms may be associated with systemic autoimmune disease, e.g., Behcet's syndrome, ankylosing spondylitis, juvenile rheumatoid arthritis, Reiter's syndrome, and inflammatory bowel disease. In anterior uveitis, inflammation is primarily in the iris (also iritis). Anterior uveitis can affect subjects who have systemic autoimmune disease, but also subjects who do not have systemic autoimmune disease. Intermediate uveitis involves inflammation of the anterior vitreous, peripheral retina, and ciliary body, often with little anterior or chorioretinal inflammation. Pan planitis results from inflammation of the pars plana between the iris and the choroid.
Posterior uveitis involves the uveal tract and primarily the choroid, and is also referred to as choroiditis. Posterior uveitis can be associated with a systemic infection or an autoimmune disease. It can persist for months and even years. A receptor binding agent can be administered to a subject to treat any of the foregoing forms of uveitis.
See also e.g., Tsai et al. (2009) Mol Vis 15:1542-1552 and Trittibach P et al. (2008)
Gene Ther. 15(22): 1478-88.
In some embodiments, a receptor binding agent is used to treat a subject having or at risk for age-related macular degeneration (AMD). The receptor binding agent can be applied topically to the eye, injected (e.g., intravitreally) or provided systemically.
See, e.g., Olson et al. (2009) Ocul Immunol Inflamm 17(3):195-200.
A receptor binding agent described herein can be administered by any mode to treat an ocular disease. The agent can be delivered by a parenteral mode. Alternatively or in addition, the agent can be delivered directly to the eye or in the vicinity of the eye. For example, the protein can be administered topically or intraocularly, e.g., as described below.
Formulations and Methods for Ocular Delivery
Ophthalmic formulations containing a receptor binding agent can be delivered for topical administration, e.g., for administration as a liquid drop or an ointment, or for implantation, e.g., into an anterior chamber of the eye or the conjunctival sac. Liquid drops can be delivered using an eye dropper. When formulated for ocular delivery, the receptor binding agent can be present at 0.0001 - 0.1 %, 0.001 -5%, e.g., 0.005-0.5%, 0.05 - 0.5%, 0.01 -5%, 0.1 -2% or 1 %-5% concentration. Frequently the ophthalmic formulation is applied directly to the eye including topical application to the eyelids or instillation into the space (cul-de-sac) between the eyeball and the eyelids. The ophthalmic formulation can be designed to mix readily with the lacrimal fluids and spread over the surfaces of the cornea and conjunctiva. With the usual technique of
administration, the major portion of the drug is deposited in the lower fornix. Capillarity, diffusional forces, and the blinking reflex drive incorporation of the drug in the precorneal film from which it penetrates into and through the cornea.
Ophthalmic formulations can also include one or more other agents, e.g., an antiinflammatory steroid such as rimexolone, loteprednol, medrysone and hydrocortisone, or a non-steroidal anti-inflammatory. For example, the steroid can be present at a concentration of 0.001 to 1 %. In some embodiments, no steroid is present. For example, the receptor binding agent is the only active agent in the formulation.
The formulation can also include one or more of the following components:
surfactants, tonicity agents, buffers, preservatives, co-solvents and viscosity building agents. Tonicity agents can be used to adjust the tonicity of the composition, e.g., to that of natural tears. For example, potassium chloride, sodium chloride, magnesium chloride, calcium chloride, dextrose and/or mannitol may be added to achieve an appropriate tonicity, e.g., physiological tonicity. Tonicity agents can be added in an amount sufficient to provide an osmolality of about 150-450 mOsm or 250-350 mOsm.
The formulation can also include buffering suitable for ophthalmic delivery. The buffer can include one or more buffering components (e.g., sodium phosphate, sodium acetate, sodium citrate, sodium borate or boric acid) to changes in pH especially under storage conditions. For example, the buffer can be selected to provide a target pH within the range of pH 6.0-7.5, e.g., 6.5-7.5.
The formulation can include an aqueous or phospholipid carrier. Particularly for treating dry eye disorders, the formulation can include agents to provide short-term relief, e.g., compounds which lubricate the eye and assist in tear formation. For example, phospholipid carriers (which include one or more phospholipids) can be used to provide short-term relief. Examples or artificial tears compositions useful as artificial tears carriers include commercial products such as Tears Naturale™ (Alcon Labs, Inc., TX USA). For example, per ml, the formulation can include: 1 mg dextran, 70 and 3 mg hydroxypropyl methylcellulose, and optionally a preservative such POLYQUAD®
(polyquaternium-1 ) 0.001 % (m/v). Examples of phospholipid carrier formulations include those disclosed in US 4,804,539, US 4,883,658, US 5,075,104, US 5,278,151 , and US 5,578,586.
The formulation can also include other compounds that act as a lubricant or wetting agent. These include viscosity agents such as: monomeric polyols, such as, glycerol, propylene glycol, ethylene glycol; polymeric polyols, such as polyethylene glycol, various polymers of the cellulose family: hydroxypropylmethyl cellulose ("HPMC"), carboxy methylcellulose sodium, hydroxy propylcellulose ("HPC"), dextrans, such as dextran 70; water soluble proteins, such as gelatin; and vinyl polymers, such as polyvinyl alcohol, polyvinylpyrrolidone, povidone and carbomers, such as carbomer 934P, carbomer 941 ; carbomer 940, carbomer 974P. Still additional examples include polysaccharides, such as hyaluronic acid and its salts and chondroitin sulfate and its salts, and acrylic acid polymers. In certain embodiments, the formulation has a viscosity between 1 to 400 cP.
The formulation can be packaged for single or multi-dose use, e.g., in a bottle with an associated dropper or as a set of single-use droppers. The formulation can include one or more preservatives, e.g., to prevent microbial and fungal contamination during use. Exemplary preservatives include: benzalkonium chloride, chlorobutanol, benzododecinium bromide, methyl paraben, propyl paraben, phenylethyl alcohol, edetate disodium, sorbic acid, and polyquaternium-1 , and can be included at a concentration of from 0.001 to 1 .0% w/v. It is also possible to provide a formulation containing a receptor binding agent that is sterile yet free of preservatives. The formulation can be prepared for single use application.
Ophthalmic packs may be used to give prolonged contact of an ophthalmic formulation with the eye. A cotton pledget is saturated with the formulation and then inserted into the superior or inferior fornix. A receptor binding agent may also be administered by the way of iontophoresis. This procedure keeps the solution in contact with the cornea in an eyecup bearing an electrode. Diffusion of the drug is effected by difference of electrical potential.
A receptor binding agent may also be delivered by injection, e.g., subconjunctival injection. The formulation can be injected underneath the conjunctiva facilitating passage through the sclera and into the eye by simple diffusion. The formulation can also be injected underneath the conjunctiva and the underlying Tenon's capsule in the more posterior portion of the eye to deliver the agent to the ciliary body, choroid, and retina. The formulation may also be administered by retrobulbar injection.
With respect to dry eye and other surface disorders, subjects can be evaluated using one or more of the following approaches: the Ocular Surface Disease Index (OSDI), corneal and conjunctival staining, and the Schirmer test.
The Ocular Surface Disease Index (OSDI) is a 12-item questionnaire that provides a rapid assessment of the symptoms of ocular irritation consistent with ocular surface inflammatory disorders, including DES, and their impact on vision-related functioning. See e.g. Ocul Immunol Inflamm. 2007 Sep-Oct;15(5):389-93. The 12 items of the OSDI questionnaire are graded on a scale of 0 to 4. Scores are derived based on responses to provide an OSDI score on a scale of 0 to 100, with higher scores representing greater disability. A negative change from baseline indicates an
improvement in vision-related function and the ocular inflammatory disorders. Corneal and Conjunctival Staining: Corneal staining is a measure of epithelial disease, or break in the epithelial barrier of the ocular surface, typically seen with ocular surface inflammatory disorders such as dry eye. Corneal staining can exist even without clinically evident dry eye, if there is significant lid disease, such as posterior blepharitis. Corneal staining is highly correlated with ocular discomfort in many, though not all patients; in general corneal staining is associated with high scores in the OSDI, as described above. For corneal fluorescein staining, saline-moistened fluorescein strips or 1 % sodium fluorescein solution are used to stain the tear film. The entire cornea is then examined using slit-lamp evaluation with a yellow barrier filter (#12 Wratten) and cobalt blue illumination. Staining is graded according to the Oxford Schema. Conjunctival staining is likewise a measure of epithelial disease or break in the epithelial barrier of the ocular surface. Conjunctival staining is performed under the slit-lamp using lissamine green. Saline-moistened strip or 1 % lissamine green solution is used to stain the tear film, and interpalpebral conjunctival staining is evaluated more than 30 seconds but less than 2 minutes later. Using white light of moderate intensity, only the interpalpebral region of the nasal and temporal conjunctival staining is graded using the Oxford Schema.
Schirmer Test: The Schirmer test is performed in the presence and in the absence of anesthesia by placing a narrow filter-paper strip (5 x 3 5mm strip of
Whatman #41 filter paper) in the inferior cul-de-sac. This test is conducted in a dimly lit room. The patient gently closes his/her eyes until five minutes have elapsed and the strips are removed. Because the tear front will continue advancing a few millimeters after it has been removed from the eyes, the tear front is marked with a ball-point pen at precisely five minutes. Aqueous tear production is measured by the length in millimeters that the strip wets during 5 minutes. Results of 10 mm or less for the Schirmer test without anesthesia and 5 mm or less for the Schirmer test with anesthesia are considered abnormal. A positive change from baseline indicates improvement of one or more symptoms of an ocular inflammatory disorder described herein.
Formulations and Methods for Pulmonary Delivery
A receptor binding agent can be formulated for inhalatory or other mode of pulmonary delivery, e.g., to administer the agent to a tissue of the respiratory tract, e.g., the upper and lower respiratory tract. The three common systems that can be used to deliver agents locally to the pulmonary air passages include dry powder inhalers (DPIs), metered dose inhalers (MDIs) and nebulizers. MDIs may be used to deliver receptor binding agents in a solubilized form or as a dispersion. Typically MDIs include a freon or other relatively high vapor pressure propellant that forces aerosolized medication into the respiratory tract upon activation of the device. In contrast DPIs generally rely on the inspiratory efforts of the patient to introduce a medicament in a dry powder form to the lungs. Nebulizers form a medicament aerosol to be inhaled by imparting energy to a liquid solution. The agent can be stored in a lyophilized form (e.g., at room temperature) and reconstituted in solution prior to inhalation.
Direct pulmonary delivery of drugs during liquid ventilation or pulmonary lavage using a fluorochemical medium are also possible delivery modes. These and other methods can be used to deliver receptor binding agent. For example, the agent is delivered in a dosage unit form of at least about 0.02, 0.1 , 0.5, 1 , 1 .5, 2, 5, 10, 20, 40, or 50 mg/puff or more.
The receptor binding agent may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane,
dielilorotetrafluoroctliane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges for use in an inhaler or insufflator may be formulated containing a powder mix of the receptor binding agent and a suitable powder base such as lactose or starch, if the particle is a formulated particle. In addition to the formulated or unformulated receptor binding agent, other materials such as 100% DPPC or other surfactants can be mixed together to promote the delivery and dispersion of the formulated or unformulated receptor binding agent. Particle size can also be varied to control whether deliver is to the lower or upper respiratory tract. For example, particles in the size range of 1 -5 microns or 10-50 microns can be used for the lower and upper respiratory tracts respectively.
Delivery enhancers such as surfactants can be used to further enhance pulmonary delivery. A surfactant is generally a compound having a hydrophilic and lipophilic moiety, which promotes absorption of a drug by interacting with an interface between two immiscible phases. Surfactants are useful in the dry particles for several reasons, e.g., reduction of particle agglomeration and reduction of macrophage phagocytosis. Surfactants are well known in the art and include phosphoglycerides, e.g., phosphatidylcholines, L-alpha-phosphatidylcholine dipalmitoyl (DPPC) and diphosphatidyl glycerol (DPPG); hexadecanol; fatty acids; polyethylene glycol (PEG); polyoxyethylene-9-; auryl ether; palmitic acid; oleic acid; sorbitan trioleate (Span 85); glycocholate; surfactin; poloxomer; sorbitan fatty acid ester; sorbitan trioleate; tyloxapol; and phospholipids.
Also featured herein are antibodies that specifically recognize a chimeric cytokine domain described herein. For example, such antibodies preferentially bind to a chimeric domain relative to any parental cytokine domain. For example, a specific antibody can bind to an epitope that includes a junction between a segment from a first parental cytokine and a second parental cytokine. Nucleic Acid Delivery
A receptor-binding agent can be provided to a subject by delivering a nucleic acid that encodes and can express the receptor-binding agent. For example, a nucleic acid sequence encoding the receptor-binding agent can be placed under control of transcription control sequences and positioned in a nucleic acid vector for gene delivery, e.g., a viral vector. Exemplary viral vectors include adenoviral, retroviral, or adeno- associated viral vectors. Vectors can be in the form of a plasmid or linear molecule, e.g., a linear double stranded DNA. The delivered nucleic acid can be designed to be incorporated into the genome of the target cell, e.g., to integrate into the genome of the target cell. Alternatively, the delivered nucleic acid can be designed such that after delivery it exists autonomously in the cell.
Transcriptional control sequences can be engineered to provide transient or constitutive expression. Transient control can include control regulated by an
exogenous agent, e.g., by using transcriptional response elements for transcription factors responsive to exogenous agents (e.g., a steroid hormone or FK506) or environmental signals.
Expression of genes provided on the delivered nucleic acid can be evaluated, e.g., by detection of the protein encoded by the gene (e.g., using antibodies) or by detection of the mRNA, e.g., using PCR or Northern hybridization. The delivered nucleic acid is generally engineered so that transcriptional and translational regulatory DNA are appropriately positioned relative to the coding sequence for the receptor-binding agent such that transcription is initiated and that protein is translated from the resulting message. The nucleic acid can include transcriptional and translational regulatory nucleic acid sequences from mammalian cells, particularly humans. Such sequences include, e.g., promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, and enhancer or activator sequences.
In addition, the expression vector may include additional elements. For example, for integrating expression vectors, the expression vector can contain at least one or two sequences homologous to the host cell genome, e.g., flanking the expression construct. The integrating vector may be directed to a specific locus in the host cell by selecting the appropriate homologous sequence for inclusion in the vector. Constructs for integrating vectors are well known in the art.
Exemplary adenoviral vectors include modified versions of human adenoviruses such as Ad2 or Ad5, in which the genetic elements necessary for the virus to replicate in vivo have been removed. For example, the E1 region can be removed and the genome can be further modified to accept an expression cassette coding for the receptor-binding agent.
Exemplary retroviral vectors include LNL6, LXSN, LNCX, and lentiviral vectors.
Particular lentiviral vectors are described by Pawliuk et al. (2001 ) Science 294:2368 and Imren et al. (2002) PNAS 99:14380 and include limited to, human immunodeficiency virus (e.g., HIV-1 , HIV-2), feline immunodeficiency virus (FIV), simian immunodeficiency virus (SIV), bovine immunodeficiency virus (BIV), and equine infectious anemia virus (EIAV). These vectors can be constructed and engineered to be safe, e.g., by separating the essential genes (e.g., gag and pol) onto separate vectors and by rendering retrovirus replication defective. The replication defective retrovirus is then packaged into virions through the use of a helper virus or a packaging cell line, by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel, F. M. et al. (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals. The retroviral vector can include, in addition to sequences for expressing a receptor-binding agent, a left (5') retroviral LTR; a retroviral export element, optionally a reverse response element (RRE); a promoter, and a locus control region (LCR) or other transcriptional insulator sequence and a right (3') retroviral LTR. Retroviral vectors can further contain a central polypurine tract (cPPT) or DNA flap to increase viral titers and transduction efficiency.
The nucleic acid containing a sequence encoding a receptor-binding agent can be delivered to any appropriate target cells, e.g., ex vivo or in vivo. Exemplary target cells include synovial cells, hematopoietic cells, dermal cells, and so forth. The nucleic acid can be delivered to target cells associated with the eye, e.g., corneal epithelial cells. Delivery may include the debridement, or scraping of the corneal epithelium to expose a basal layer of epithelium. The nucleic acid for delivery is then added. In another embodiment, the nucleic acid is delivered to corneal endothelial cells, cells of the trabecular meshwork beneath the periphery of the cornea, cells of the choroid layer of the eye, cells of the retina, sclera or ciliary body, cells of the retinal or ocular vasculature, or cells of the vitreous body or cells of the lens, for example the lens epithelium.
Delivery methods include, e.g., retroviral infection, adenoviral infection, transformation with plasmids, transformation with liposomes containing exogenous nucleic acid, biolistic nucleic acid delivery (e.g., loading the nucleic acid onto gold or other metal particles and shooting or injecting into the cells), adeno-associated virus infection and Epstein-Barr virus infection. Delivery can be into cells or tissue by any method including needle injection, hypospray, electroporation, or a gene gun.
Other methods for gene delivery can be found in, e.g., Kay, M. A. (1997) Chest
1 1 1 (6 Supp.):138S-142S; Ferry, N. and Heard, J. M. (1998) Hum. Gene Ther. 9:1975- 81 ; Shiratory, Y. et al. (1999) Liver 19:265-74; Oka, K. et al. (2000) Curr. Opin. Lipidol. 1 1 :179-86; Thule, P. M. and Liu, J. M. (2000) Gene Ther. 7:1744-52; Yang, N. S. (1992) Crit. Rev. Biotechnol. 12:335-56; Alt, M. (1995) J. Hepatol. 23:746-58; Brody, S. L. and Crystal, R. G. (1994) Ann. N.Y. Acad. Sci. 716:90-101 ; Strayer, D. S. (1999) Expert Opin. Investig. Drugs 8:2159-2172; Smith-Arica, J. R. and Bartlett, J. S. (2001 ) Curr. Cardiol. Rep. 3:43-49; and Lee, H. C. et al. (2000) Nature 408:483-8.
Exemplary Second Agents
A receptor binding agent described herein can be administered with a second agent. The two agents can be co-administered, or administered separately, e.g., using different regimes. Exemplary second agents include an anti-inflammatory agent.
In one embodiment, the second agent is an IL-17 antagonist (encompassing antagonists of all IL-17 family members, e.g., antagonists of IL-17A, IL-17F, IL-17B, IL-17C, IL-17D, and IL-17E). Exemplary IL-17 antagonists include: agents (such as antibodies and other binding proteins) that bind to IL-17 (including IL-17A, IL-17F, IL-17B, IL-17C, IL-17D, and IL-17E) and which antagonize IL-17 mediated signaling; agents (such as antibodies and other binding proteins) that bind to one or more receptors for IL-17, such as IL-17RA and IL-17RC and which antagonize IL-17 mediated signaling; agents (such as antibodies and other binding proteins) that bind to a complex containing IL-17 and at least one receptor subunit, e.g., IL-17 and 11-17RA, or IL-17, IL-17RA, and IL-17RC and which antagonize IL-17 mediated signaling; and agents such as soluble receptors that include one or more of soluble extracellular domains of IL-17RA and IL-17RC and which antagonize IL-17 mediated signaling.
In another embodiment, the second agent is an IL-12 antagonist. Exemplary IL- 12 antagonists include: agents (such as antibodies and other binding proteins) that bind to IL-12 (including p35 and p40) and which antagonize IL-12 mediated signaling; agents (such as antibodies and other binding proteins) that bind to one or more receptors for IL- 12, such as IL-12R 1 or IL-12R32 and which antagonize IL-12 mediated signaling; agents (such as antibodies and other binding proteins) that bind to a complex containing p35, p40 and at least one receptor subunit, e.g., IL-12R 1 or IL-12R32 and which antagonize IL-12 mediated signaling; and agents such as soluble receptors that include one or more of soluble extracellular domains of IL-12R31 or IL-12R32 and which antagonize IL-12 mediated signaling.
In another embodiment, the second agent is an IL-23 antagonist. Exemplary IL- 23 antagonists include: agents (such as antibodies and other binding proteins) that bind to IL-23 (including p19 and p40) and which antagonize IL-23 mediated signaling; agents (such as antibodies and other binding proteins) that bind to one or more receptors for IL- 23, such as IL-12R 1 or IL-23R and which antagonize IL-23 mediated signaling; agents (such as antibodies and other binding proteins) that bind to a complex containing p19, p40 and at least one receptor subunit, e.g., IL-12R i or IL-23R and which antagonize IL- 23 mediated signaling; and agents such as soluble receptors that include one or more of soluble extracellular domains of IL-12R 1 or IL-23R and which antagonize IL-23 mediated signaling.
Exemplary antibodies to IL-23 have been described. See e.g., Beyer et al., J. Mol. Biol. (2008), doi:10.1016/j.jmb.2008.08.001 .
Animal Models
A receptor binding agent can be evaluated in an animal model for human disease, e.g., a human autoimmune and/or human inflammatory disease. The agent can have specificity for the corresponding target protein in the animal.
Rheumatoid Arthritis Models. A receptor binding agent can be evaluated in an animal model of rheumatoid arthritis, e.g., the collagen-induced arthritis (CIA) model. See, for example, Mclndoe et al., 1999, Proc. Natl. Acad. Sci. USA, 96:2210-2214; Issekutz, A. C. et al., Immunology (1996) 88:569; and Current Protocols in Immunology, Unit 15.5, Coligan et al. (eds.), John Wiley & Sons, Inc. The model is produced by the immunization of susceptible strains of rat/mice with native type II collagen. Collagen is emulsified in Complete Freund's Adjuvant (CFA) and injected intradermal^ (100 μg collagen:100 μg CFA/mouse) at the base of the tail. Control mice are injected intradermal^ with 0.05 ml of distilled water/CFA emulsion. A booster injection of collagen in incomplete adjuvant is given 21 days after the initial immunization. Disease is due to an autoimmune response induced upon immunization with collagen.
The joints can be scored for arthritis, inflammation, pannus, cartilage damage and bone resorption using a defined scale. For example the severity of arthritis can be scored as follows: 0=no visible effects of arthritis; 1 =edema and erythema of one digit or joint; 2=edema and erythema of two joints; 3=edema and erythema of more than two joint; 4=severe arthritis of the entire paw and digits, accompanied by ankylosis of the ankle and deformity of the limb. The score for each limb is summed and recorded as the arthritic index (Al) for each individual animal. Other scoring schemes can also be used for these and other criteria.
Multiple Sclerosis. Experimental allergic encephalomyelitis (EAE) is a useful murine model for multiple sclerosis. A receptor binding agent can be evaluated in the EAE model. EAE is a T cell mediated autoimmune disorder characterized by T cell and mononuclear cell inflammation and subsequent demyelination of axons in the central nervous system. (See, for example, Bolton, C, 1995, Multiple Sclerosis, 143.)
Exemplary protocols can be found in Current Protocols in Immunology, Unit 15.1 and
15.2; Coligan et al. (eds.), John Wiley & Sons, Inc. Models are also available for myelin disease in which oligodendrocytes or Schwann cells are grafted into the central nervous system, for example, as described in Duncan et al., 1997, Molec. Med. Today, 554-561 .
Allograft. A receptor binding agent can be evaluated in an animal model for skin allograft rejection, e.g., using murine tail-skin grafts. Skin allograft rejection is mediated by T cells, helper T cells and killer-effector T cells. See, for example, Current Protocols in Immunology, Unit 4.4; Coligan et al. (eds.), 1995, John Wiley & Sons, Inc. Other transplant rejection models can also be used. See, e.g., Tinubu et al., 1994, J.
Immunol., 4330-4338. IBD and Colitis Models. An exemplary model for inflammatory bowel disease is the use of CD4+ CD45Rb-high cells transferred to SCID mice. See e.g., Hirano et al., J Pharmacol Sci. 2009 Jun;1 10(2):169-81 and the use of transgenic IL-10 deficient mice. See e.g., Inaba et al., Inflamm Bowel Dis., DOI: 10.1002/ibd.21253 (2010). Yet another exemplary colitis model employs dextran sulfate sodium (DSS) to induce acute colitis. For example, colitis can be induced in mice by administration of 5% (wt/vol) DSS (molecular mass 30-40 kDa; ICN Biomedicals, Aurora, OH) in drinking water ad libitum. Symptoms resulting from this treatment bloody diarrhea, weight loss, colon shortening and mucosal ulceration with neutrophil infiltration. DSS-induced colitis is characterized histologically by infiltration of inflammatory cells into the lamina propria, with lymphoid hyperplasia, focal crypt damage, and epithelial ulceration. These changes are thought to develop due to a toxic effect of DSS on the epithelium and by phagocytosis of lamina propria cells and production of TNF-alpha and IFN-gamma. See, e.g., Hassan et al. PLoS One. 2010 Jan 25;5(1 ):e8868.
Dry Eye Disease Models. A receptor binding agent can be evaluated in a mouse model for dry eye disease. Dry eye can be induced in mice by subcutaneous injection of scopolamine and then placement of the mice in controlled-environment chambers. By way of a specific example, normal healthy 6 to 10 weeks old female C57BL/6 mice can be induced to have dry eye by continuous exposure to dry environment in a controlled environmental chamber. The chamber has low relative humidity of less than 30% (generally about 19%), high airflow (15 liters/minute) and constant temperature (about 22 <C). The mice placed in the chamber are also treated with scopolamine to inhibit tear secretion. Sustained-release transdermal scopolamine patches can be obtained from Novartis (Summit, N.J.). One-fourth of a patch is applied to the depilated mid-tail of mice every 48 hours. The combination of the controlled environmental chamber and scopolamine produces severe dry eye in a relative short period of time (about 2-4 days). The controlled environmental chamber can be prepared as described in Barbino et al., Invest. Ophthal. Vis. Sci., 46: 2766-271 1 (2005), and enables control of air flow, humidity, and temperature.
Mice can be monitored for signs of dry eye, e.g., by performing: a) cotton thread test to measure aqueous tear production, which is generally decreased in patients with dry eye; b) corneal fluorescein staining which is a marker of corneal surface damage; and general ophthalmic examination. Cotton Thread Test: Tear production can be measured with cotton thread test, impregnated with phenol red (Zone-Quick, Lacrimedics, Eastsound, Wash.). Under a magnifying fluorescent lamp, the thread is held with jeweler forceps and placed in the lateral cantus of the conjunctival fornix of the right eye for 30 or 60 seconds. The tear distance in mm is read under a microscope using the scale of a hemacytometer.
Corneal Fluorescein Staining: Corneal fluorescein staining can be evaluated by applying 1 .0 μΙ of 5% fluorescein by a micropipette into the inferior conjunctival sac of the eye. The cornea is examined with a slit lamp biomicroscope using cobalt blue light 3 minutes after the fluorescein instillation. Punctuate staining is recorded in a masked fashion using a standardized National Eye Institute (NEI) grading system of 0-3 for each of the five areas in which the corneal surface has been divided.
Diagnostic and Other Uses
A receptor binding agent described herein can be used to detect IL-1 R1 in a sample, or cells expressing such a receptor. For example, the agent can be labeled directly or indirectly with a moiety that is a label or produces a signal, e.g., an enzyme, a radiolabel, an epitope, or a fluorescent protein (such as green fluorescent protein). The agent can be contacted to a sample or to cells to determine if the receptor is present in the sample or on the cells, e.g., using standard immunoblotting, immunofluorescence, enzyme immunoassay (EIA), radioimmunoassay (RIA), fluorescence energy transfer, Western blot, and other diagnostic and detection techniques.
The receptor binding agent can also be labeled for in vivo detection and administered to a subject. The subject can be imaged, e.g., by NMR or other tomographic means. For example, the binding agent can be labeled with a radiolabel such as 3 , η, 23l, 99mTc, 32P, 25l, 3H, 4C, and 88Rh, fluorescent labels such as fluorescein and rhodamine, nuclear magnetic resonance active labels, positron emitting isotopes detectable by a positron emission tomography ("PET") scanner,
chemiluminescers such as luciferin, and enzymatic markers such as peroxidase or phosphatase. The agent can be labeled with a contrast agent such as paramagnetic agents and ferromagnetic or superparamagnetic (which primarily alter T2 response) A receptor binding agent can also be used to purify cells which express the receptor to which it binds. For example, the receptor binding agent can be coupled to an immobilized support (e.g., magnetic beads or a column matrix) and contacted to cells which may express the receptor. The support can be washed, e.g., with a physiological buffer, and the cells can be recovered from the support.
A receptor binding agent can also be used to purify soluble forms of the receptor to which it binds. For example, samples containing the soluble receptor can be contacted to immobilized receptor binding agent and then, e.g., after washing, can be recovered from the immobilized agent.
The following non-limiting examples further illustrate embodiments of the inventions described herein.
Examples
Example 1
Nucleic acids encoding the proteins with the amino acid sequences listed in Table 4 (below) were constructed in a pET vector containing a T7 promoter and ampicillin (pET31 series) or kanamycin resistance genes (pET28 series) (EMD
Chemicals, Gibbstown NJ, USA), and expressed. Examples of coding sequences that can be used for expression are provided in Table 5.
Table 4
Figure imgf000078_0001
P05 APVRSLNCRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMSFVQGE 21 ESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEKRF VFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGVMVTKFY MQFVSS
P06 APVRSLNCTLWDVNQKTFYLRNNQLVAGYLQGPNVEQQVVFSMSFVQGE 23
ESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEKRF VFNKIEI NKLEFESAQFPNWYI STSMEADQPVFLGGTKGGQDITDFTM QFVSS
P07 APVRSLNCRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMSFVQGE 24
ESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEKRF VFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGQDITDFT MQFVSS
Exemplary nucleic acid sequences encoding the above proteins are listed in Table 5. In some embodiments, the nucleic acid sequence further includes an ATG prior to the first nucleotide listed below. In some embodiments, the nucleic acid sequence further includes a stop codon (such as TAA, TAG, or TGA) after the last nucleotide listed below.
Table 5
Figure imgf000079_0001
P03 GCACCTGTACGATCACTGGCCTTCAGAATCTGGGATGTTAACCAGAAGA 27 CCTTCTATCTGAGGAACAACCAACTAGTTGCTGGATACTTGCAAGGACC AAATGTCAATTTAGAAGAAAAGTTCTCCATGTCCTTTGTACAAGGAGAA GAAAGTAATGACAAAATACCTGTGGCCTTGGGCCTCAAGGAAAAGAATC TGTACCTGTCCTGCGTGTTGAAAGATGATAAGCCCACTCTACAGCTGGA GAGTGTAGATCCCAAAAATTACCCAAAGAAGAAGATGGAAAAGCGATTT GTCTTCATCCGCTCAGACAGCGGCCCCACCACCAGTTTTGAGTCTGCCG CCTGCCCCGGTTGGTTCCTCTGCACAGCGATGGAAGCTGACCAGCCCGT CAGCCTCACCAATATGCCTGACGAAGGCGTCATGGTCACCAAATTCACC ATGCAATTTGTGTCTTCC
P04 GCACCTGTACGATCACTGGCCTTCAGAATCTGGGATGTTAACCAGAAGA 28
CCTTCTATCTGAGGAACAACCAACTAGTTGCTGGATACTTGCAAGGACC AAATGTCAATTTAGAAGAAAAGTTCTCCATGTCCTTTGTACAAGGAGAA GAAAGTAATGACAAAATACCTGTGGCCTTGGGCCTCAAGGAAAAGAATC TGTACCTGTCCTGCGTGTTGAAAGATGATAAGCCCACTCTACAGCTGGA GAGTGTAGATCCCAAAAATTACCCAAAGAAGAAGATGGAAAAGCGATTT GTCTTCAACAAGATAGAAATCAATAACAAGCTGGAATTTGAGTCTGCCG CCTGCCCCGGTTGGTTCCTCTGCACAGCGATGGAAGCTGACCAGCCCGT CAGCCTCACCAATATGCCTGACGAAGGCGTCATGGTCACCAAATTCACC ATGCAATTTGTGTCTTCC
P05 GCACCTGTACGATCACTGAACTGCAGAATCTGGGATGTTAACCAGAAGA 29
CCTTCTATCTGAGGAACAACCAACTAGTTGCTGGATACTTGCAAGGACC AAATGTCAATTTAGAAGAAAAGTTCTCCATGTCCTTTGTACAAGGAGAA GAAAGTAATGACAAAATACCTGTGGCCTTGGGCCTCAAGGAAAAGAATC TGTACCTGTCCTGCGTGTTGAAAGATGATAAGCCCACTCTACAGCTGGA GAGTGTAGATCCCAAAAATTACCCAAAGAAGAAGATGGAAAAGCGATTT GTCTTCAACAAGATAGAAATCAATAACAAGCTGGAATTTGAGTCTGCCC AGTTCCCCAACTGGTTCCTCTGCACAGCGATGGAAGCTGACCAGCCCGT CAGCCTCACCAATATGCCTGACGAAGGCGTCATGGTCACCAAATTCTAC ATGCAATTTGTGTCTTCC
P06 GCACCTGTACGATCACTGAACTGCACGCTCTGGGATGTTAACCAGAAGA 30
CCTTCTATCTGAGGAACAACCAACTAGTTGCTGGATACTTGCAAGGACC AAATGTCGAGCAACAAGTGGTGTTCTCCATGTCCTTTGTACAAGGAGAA GAAAGTAATGACAAAATACCTGTGGCCTTGGGCCTCAAGGAAAAGAATC TGTACCTGTCCTGCGTGTTGAAAGATGATAAGCCCACTCTACAGCTGGA GAGTGTAGATCCCAAAAATTACCCAAAGAAGAAGATGGAAAAGCGATTT GTCTTCAACAAGATAGAAATCAATAACAAGCTGGAATTTGAGTCTGCCC AGTTCCCCAACTGGTACATCAGCACCTCTATGGAAGCTGACCAGCCCGT CTTCCTGGGAGGGACCAAAGGCGGCCAGGATATAACTGACTTCACCATG CAATTTGTGTCTTCC P07 GCACCTGTACGATCACTGAACTGCAGAATCTGGGATGTTAACCAGAAGA 31
CCTTCTATCTGAGGAACAACCAACTAGTTGCTGGATACTTGCAAGGACC
AAATGTCAATTTAGAAGAAAAGTTCTCCATGTCCTTTGTACAAGGAGAA
GAAAGTAATGACAAAATACCTGTGGCCTTGGGCCTCAAGGAAAAGAATC
TGTACCTGTCCTGCGTGTTGAAAGATGATAAGCCCACTCTACAGCTGGA
GAGTGTAGATCCCAAAAATTACCCAAAGAAGAAGATGGAAAAGCGATTT
GTCTTCAACAAGATAGAAATCAATAACAAGCTGGAATTTGAGTCTGCCC
AGTTCCCCAACTGGTTCCTCTGCACAGCGATGGAAGCTGACCAGCCCGT
CAGCCTCACCAATATGCCTGACGAAGGCCAGGATATAACTGACTTCACC
ATGCAATTTGTGTCTTCC
The proteins can include a range of different residues from IL-1 β and IL-1 Ra as illustrated below. Among the examples P01 , P02, P03, P04, and P05, the cytokine domains can have 48-70% residues from I L- 1 β and 55-78% residues from IL-1 Ra. (Because a number of amino acid residues are conserved between the two proteins, the sum of the percentage identity to IL-1 β and to IL-1 Ra can be greater than 100%.) Table 6
Figure imgf000081_0001
Example 2
Proteins that contain a hexa-histidine tag were expressed in E. coli cells
BL21 (DES) strain by induction with 1 mM IPTG at 37°C for 3 hours in LB broth media. The cells were lysed in 20-50 mM Tris, 0.5 M NaCI, 2.5 mM EDTA, 0.1 % Triton X-100, pH 8.0. Lysates were subjected to IMAC chromatography using a HiTrap® pre-packed column (GE Healthcare, Piscataway NJ, USA). The protein was loaded in 20 mM sodium phosphate, 0.5 M NaCI 10 mM imidazole, pH 7.4 buffer. It was eluted with 200 mM imidazole, 20 mM sodium phosphate, 0.5 M NaCI pH 7.4 buffer. Eluted protein was dialyzed extensively against PBS, 0.1 % Polysorbate 80, pH 7.4, concentrated using an Amicon Ultra® (1 OK) filter, and stored at 4° or -80 °C. Proteins lacking a hexa-histidine tag were purified by ion exchange chromatography. P05 protein was purified by ion exchange chromatography. Lysate from expressing cells was applied to a GigaCapS™ column (Tosoh Bioscience LLC, King of Prussia, PA, USA) at low pH (approximately pH 5.5) in the absence of salt (conductivity approximately 1 mS/cm). The column was then eluted by a pH gradient (Buffer A = 10 mM acetic acid, pH 5.5; Buffer B = 20 mM Tris pH 8). A 5 ml fraction containing the eluted protein was then diluted with 5 ml of H20 and 5 ml of 20 mM Tris pH 8) and then applied to CaptoQ™ resin (GE Healthcare, Piscataway NJ, USA) and eluted with a 0 mM to 250 mM NaCI gradient in 20 mM Tris pH 8.0. The eluted protein was dialyzed extensively against 1 .25 X PBS 0.1 % TWEEN® 80 or 1 .25X PBS lacking TWEEN® and stored. See Fig. 6. P03 and P04 proteins were purified using similar methods.
Cells expressing P05 were also grown in TEKNOVA™ Terrific Broth with animal free soytone (# T7660) supplemented with 10 g/L glucose, 10 mM MgS04, trace elements (1 mg/ml TEKNOVA™ 1000X Trace Elements, #T1001 ), and antibiotic in a Sartorius 2L BIOSTAT™ A+ and were induced at OD 35-40 with 1 mM IPTG for about 6 hours. Cells are grown at 37°C with 30% dissolved oxygen at pH 7.0, and agitation at 200-800 rpm with oxygen sparge at 2L/min. Cells are fed 9 g glucose/L/hr when glucose is depleted as detected by a pH increase. Feed is reduced to 6 g glucose/L/hr when the pH decreases (about 2.5 hrs after induction).
Cells were collected and lysed in lysis buffer (20 mM Tris, 10 mM EDTA, 0.1 % Triton, pH 8.0; 20 mM Tris, 10 mM EDTA, 0.1 % Triton, pH 7.0; 50 mM MOPS, 10 mM EDTA, 0.1 % Triton, pH 6.5; or 50 mM MOPS, 10 mM EDTA, 0.1 % Triton, pH 6.0).
Lysate is loaded onto Poros XS® cation ion exchange media (Life Technologies Corp., Carlsbad CA USA) at pH 5.3 and 3 mS/cm (35 mg product per ml column resin).
In an exemplary procedure, P05 protein is eluted by a step to pH 7.0 using buffer containing 100 mM MOPS 25 mM NaCI pH 7.0. The first eluting peak was discarded, and the second eluting peak was collected in pools and contained P05 protein. Early pools are enriched for intact P05 protein relative to a des-Ala species. This eluted material is then flowed over CaptoQ™ anion exchange resin. The flow through, which contains intact P05 protein, is collected.
In another exemplary procedure, the media is washed with 100 mM MOPS 20 mM NaCI pH 6.0. P05 protein is eluted by a step to pH 6.0 using buffer containing 100 mM MOPS 50-58 mM NaCI pH 6.0. The first eluting peak was separated from subsequent peaks and contained intact P05 protein. This eluted material is then flowed over CaptoQ™ anion exchange resin. The flow through, which contains intact P05 protein, is collected.
Example 3
The proteins or supernatants containing the proteins were evaluated in a cell- based assay for IL-1 activity. HEK-Blue™ I L-1 β responsive cells were used to monitor IL-1 β activity (available from InvivoGen Inc., San Diego CA, USA). These cells include a SEAP reporter gene under the control of the IFN-β minimal promoter fused to five NF-KB and five AP-1 binding sites. IL-1 β engagement of IL-1 receptors on the cell surface lead to NF-KB activation and SEAP production. The SEAP report can be detected, e.g., using QUANTI-Blue™ (InvivoGen Inc., San Diego CA, USA) and spectrophotometric analysis. A HEK-Blue IL-1 β cell suspension was prepared from cells cultured to 70-80% confluence. The resuspended cells were adjusted to -330,000 cells/ml in fresh growth medium (DMEM, 4.5 g/l glucose, 2 mM L-Glutamine, 10% (v/v) heat-inactivated fetal bovine serum (30 min at 56°C), 50 U/ml penicillin, 50 μg/ml streptomycin, 100 μg/ml NormocinT).
Reagents were added to wells of a flat-bottom 96-well cell culture plate: 10 μΙ of IL-1 β at 20 ng/ml, 1 μΙ of the agent of interest and 30 μΙ of cell culture medium to a final volume of 50 μΙ. Positive and negative controls samples were prepared in parallel.
Then 150 μΙ of HEK-Blue IL-1 β cell suspension (-50,000 cells) was added to each well and the plate was cultured overnight at 37°C in 5% C02 tissue culture incubator.
Generally the final IL-1 β concentration (in the 200 μΙ final volume) was 0.1 ng/ml. I L-1 β activity was evaluated the next day (12-15 hours later). Prior to quantitation, the
QUANTI-Blue™ reagent was prepared according to the manufacturer's instructions. A flat bottomed 96-well assay plate was prepared in which 150 μΙ of QUANTI-Blue™ solution was added to each well. 50 μΙ of conditioned media from the wells of the 96 well tissue culture plate was added to each well of the assay plate. The plate was incubated at 37°C for approximately 15-20 minutes. SEAP levels were then measured using a spectrophotometer at 620-655 nm.
Results. As shown in FIG. 7A, in this assay, the P06 protein behaved as an
IL-1 Rl agonist, the P07 protein behaved as a partial agonist, and the P01 protein failed to agonize. In fact, the P01 protein behaved as an antagonist when assayed in the presence of IL-1 β. FIG. 7B shows antagonism of IL-1 β activity by P01 at a range of IL-1 β protein concentrations using the HEKBlue™ cell assay above. Antagonism increased with increasing amounts of P01 (x-axis reflects microliters of supernatant containing P01 ).
The proteins P01 , P02, P03, P04, and P05 each antagonized IL-1 β activity. See FIG. 8A and 8B, for example. The IC50 of P05 was less than about 5 ng/ml. P05 was test for ability to agonize IL-1 Rl in this assay and was not observed to have any detectable agonistic activity even at the highest concentrations tested, 1 mg/ml. P01 , P02, P03, P04, and P05 also inhibited IL-1 β induced IL-6 expression in MG-63 cells, a human osteosarcoma cell line that is responsive to IL-1 β. In a murine model of dry eye disease, hexa-histidine tagged P05 was observed to have biological activity. See also Example 8 below regarding untagged P05.
Example 4
The binding properties of proteins for soluble recombinant human IL-1 Rl (corresponding to the extracellular domain of IL-1 Rl) were evaluated using surface plasmon resonance with a Reichert SR7000DC Dual Channel SPR system. Binding was evaluated in phosphate buffered saline with 0.005% Tween 20. IL-1 β was observed to have a KD of between 8-9 nM and a dissociation constant (Kd) of between 2-3 χ 1 0"3 s~\ and in another experiment a KD of about 2 nM, an association constant of 1 .3-1 .5 χ 106 M~V, and a dissociation constant (Kd) of about 2.9-3.0 χ 10~3 s"1. See Fig. 9A. The P01 protein bound with similar association kinetics as I L-1 β , but did not dissociate during of the dissociation phase of the binding experiment (about 1 80 seconds). Thus, the P01 protein bound to IL-1 Rl with a greater affinity than did IL-1 β under similar conditions.
Binding of IL-1 Ra was observed to have a KD of about 0.33 nM, an association constant (Ka) of about 2 χ 1 05 M~ s~\ and a dissociation constant (Kd) of about 6.6 χ 1 0~5 s~ . See Fig. 9B. Chimeric cytokine domains P01 , P02, P03, P04, and P05 were observed to have KD ranging from about 1 2-1700 pM, an association constant (Ka) ranging from about 3 χ 104 M~V to 3 χ 106 M~ s~\ and a dissociation constant (Kd) ranging from about 2 χ 10~5 to 1 χ 10~3 s~ . See for example Fig. 9C and 9D and Table 7 below. Table 7
Figure imgf000085_0001
Example 5
Additional exemplary chimeric IL-1 family proteins also include the following:
P08 APVRSLAFRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMSFVQG SEQ ID EESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEK NO : 32 RFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGVMVT KFYMQFVSS
P09 APVRSQAFRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMSFVQG SEQ ID EESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEK NO : 33 RFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGVMVT KFYMQFVSS
P10 APVRSLAFRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKIDVSFVQG SEQ ID EESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEK NO : 34 RFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGVMVT KFYMQFVSS
P1 1 APVRSLNCRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKIDVSFVQG SEQ ID EESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEK NO : 35 RFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGVMVT KFYMQFVSS
P12 APVRSLNCRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMSFVQG SEQ ID EESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEK NO : 36 RFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGVMVT KFTMQFVSS P13 APVRSLAFRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMSFVQG SEQ ID EESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEK NO : 37 RFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGVMVT KFYFQED
P14 APVRSLNCRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMSFVQG SEQ ID EESNDKIPVALGLKEKNLYLSCVLKDDKPTLQLESVDPKNYPKKKMEK NO : 38 RFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGVMVT KFYFQED
The polypeptide below is a chimeric domain that includes at least two segments from IL-1 a and at least two segments from IL-1 Ra.
SAPFSFLSNVKYNFMRI IKYEFRIWDVNQKTFYLRNNQLVAGYLQGPNVN SEQ ID
LEEKFDMGAYKSSKDDAKITVILRISKTQLYVTAQDEDQPVLLKEMPEIP NO : 39 KTITGSETNLLFFWETHGTKNYFTSVAHPNLFLCTAMEADQPVSLTNMPD EGVMVTKFYILENQA Example 6
A circularly permuted IL-1 chimeric domain can be constructed by linking the N- terminus to the C-terminus of the molecule using a linker sequence and selecting a new location for each of the termini. For proteins having termini derived from IL-1 β, the linker length can be between five to ten, e.g., about seven amino acids. Preferred locations for new termini are in loops which face away from the receptors, such as the β6-β7 loop (e.g., amino acids corresponding to 71 -80 of SEQ ID NO:1 ) or the β7-β8 loop (e.g., amino acids corresponding to 84-99 of SEQ ID NO:1 ).
Examples of such circularly permuted IL-1 chimeric domains include: DKPTLQLESVDPKNYPKKKMEKRFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDE GVMVTKFYMQFVSSGGSGGGSAPVRSLNCRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMS FVQGEESNDKIPVALGLKEKNLYLSCVLKD (SEQ ID NO: 40) and
NYPKKKMEKRFVFNKIEINNKLEFESAQFPNWFLCTAMEADQPVSLTNMPDEGVMVTKFYMQFVS SGGSGGGSAPVRSLNCRIWDVNQKTFYLRNNQLVAGYLQGPNVNLEEKFSMSFVQGEESNDKIPV ALGLKEKNLYLSCVLKDDKPTLQLESVDPK (SEQ ID NO: 41) Example 7
Proteins P03, P04, P05, mll_-1 Ra (methionyl IL-1 Ra), and IL-1 β were prepared in phosphate-buffered saline (PBS), pH 7.4, at 0.5 mg/ml. The proteins were combined with SYPRO orange dye (Invitrogen, CA) at a 1 :500 dilution of the stock concentration and subject to differential scanning fluorimetry. See, e.g., He et al. (2010) J. Pharm. Sciences, 99 1707-1720. Fluorescence measurements were monitored using an Agilent Mx3005 QPCR machine as the temperature was increased from 25°C to 95°C at a rate of 1 °C per minute. Melting temperature (Tm) values were derived from the maxima value of the first derivative of the fluorescence transition. The proteins P03, P04, and P05 were observed to have an onset of unfolding of greater than 50°C and as high as 59°C, and Tm of greater than 59, 60, 62, and 64°C. Results are shown in Table 8 below and FIG. 10A & 10B:
Table 8
Protein Tm (°C) Onset of unfolding (°C)
mll_-1 Ra 56 48
IL-1 β 56 41
P03 65 59
P04 60 51
P05 65 59 P04 has a Tm that is about 4 degrees higher than mlL-1 Ra and IL-1 β and exhibits an onset of unfolding about 3 degrees higher than mlL-1 Ra and about 10 degrees higher than IL-1 β. P03 and P05 have a Tm that is about 9 degrees higher than mlL-1 Ra and IL-1 β and exhibit an onset of unfolding about 1 1 degrees higher than mlL-1 Ra and about 18 degrees higher than IL-1 β. Example 8
Purified P05 (lacking a hexa-histidine tag) was prepared in 1 .25x PBS and tested in a murine model of dry eye disease. In this model, female C57BL/6 mice 6 to 10 weeks of age from Jackson Laboratories (acclimated 1 to 2 weeks in an animal holding room with≥30% relative humidity, hydrogel food supplement, and envirodry environment enrichment) were pre-screened for fluorescein staining on Day 0. For fluorescein staining, freshly made fluorescein diluted in WFI H20 at 10 mg/mL was administered at 0.4 μ\- to each eye. Approximately 8-13 minutes after administration, eyes were scored using an Olympus fluorescent dissecting microscope. Punctuate staining was recorded using the standardized National Eye Institute (NEI) grading system of 0-3 for each of the five areas into which the corneal surface has been divided (score range 0-15/eye). Using a teaching bridge, two masked scorers evaluated mice at the same time to give a single collective score for each eye.
Mice with scores < 7 for each eye (out of a maximal score of 15) were placed in a dry eye chamber (20%± 2% humidity and constant air flow -21 L/min/cage) on day 1 and were maintained in this chamber during the course of the experiment (except for examination). On day 3, mice were scored again and randomized into treatment groups with 8 to 10 mice/group. Mice were randomized such that each cage of 4 to 5 mice had approximately the same mean disease score. Beginning on day 3 and after
randomization, mice were topically administered P05 or vehicle (1 .25X PBS) in an eye drop at 3 μί/eye BID frequency. Mice were examined and scored on days 7, 9, and 1 1 for corneal fluorescein staining as described above. Scorers were blinded as to the treatment groups during the course of the experiment.
FIG. 1 1 A is a bar graph of mean corneal staining score ± SEM at day 0, 3, 7, 9, and 1 1 for mice from two identical experiments under the following bid treatments: no treatment, vehicle (1 .25X PBS), and 10 mg/ml (1 %) P05. 10 mg/ml P05 significantly reduced corneal staining on days 7, 9, and 1 1 of the experiment. Efficacy as evaluated by a reduction in corneal staining was also observed with doses as low as 0.1 mg/ml P05. Recombinant IL-1 Ra produced in E. coli also moderately reduced corneal staining in the animal model.
As shown in FIG. 1 1 B, the effect of 10 mg/ml P05 was specific based on a comparison to 10 mg/ml murine serum albumin in the same vehicle. No effect was seen with 10 mg/ml murine serum albumin (MSA) relative to vehicle, and the effect of 10 mg/ml P05 was statistically significant relative to 10 mg/ml murine serum albumin. As shown in FIG. 1 1 C, 10 mg/ml P05 was also compared to 0.05% cyclosporine in an ophthalmic emulsion (Restasis®). Whereas P05 reduced corneal staining, no effect was observed for the 0.05% cyclosporine ophthalmic emulsion after ~1 week of bid dosing.
Example 9
A capture step has been developed for the purification of P05 produced by fermentation in BLR(DE3) E. coli cells. The elution conditions were defined through a statistical design of experiment approach (DOE) on a 1 ml_ column. The optimized conditions were performed on an intermediate scale (10 ml_ column). The product is extracted by microfluidization in a lysis buffer consisting of 20 mM Tris at pH 7.0 with 10 mM EDTA added. The clarified lysate is adjusted to a pH of 5.3 and a conductivity of 3 mS/cm. The conditioned lysate is loaded onto a PorosXS (strong cation exchange resin) with a 2 min residence time to a capacity of 25-30 mg P05/ml_ column. The column is washed with equilibration buffer to remove unbound species. A second wash at pH 6.0 and 3 mS/cm is implemented to remove a population of impurities. The product is eluted at pH 6.0 and 6.6 mS/cm. A product related species is eluted at pH 6.0 and 12.4 mS/cm. The column is cleaned with a high salt buffer and NaOH.
P05 is a 17 kDa protein with a pi of 6.58. The pi of the des-ALA species is 6.8 and is readily resolved by analytical weak cation exchange chromatography. This species is likely to be a product of aminopeptidase P activity found in E. coli. The species can be identified by mass spectroscopy, peptide mapping, and HPLC methods.
Chromatographic Materials. PorosXS is based on a crosslinked poly(styrene- divinylbenzene) bead with a nominal 50 μηι particle size. The chromatographic matrix has sulfopropyl surface chemistry and was designed to tolerate elevated levels of salt during binding. The PorosXS (CN 4404339, Life Technologies) material was procured as a bulk slurry and packed into a 5x50 mm Tricorn column (CN 28-4064-09, GE Healthcare) with a final column volume of 1 ml_ (5 cm bed height) or into a 1 0x 1 50 mm Tricorn column (CN 28-4064-16, GE Healthcare) packed to a final volume of 10.5 ml_ (13.4 cm bed height).
Buffers. All buffers were prepared with MilliQ water volumetrically. To reach the desired pH, either 10 N HCI or 1 M NaOH (made from a stock solution of 10 M NaOH) was added to titrate the buffer. After preparation, all buffers were filtered through 0.2 μηι PES bottle top filters. All pH and conductivity measurements were performed at room temperature (~ 20-25°C).
PorosXS Buffers: CEX equilibration buffer - 10 mM acetic acid (HoAC) with 21 mM NaCI made by adding 0.57 ml_ of glacial acetic acid and 2.44 g of NaCI per L of buffer. The final pH was 5.3 and the final conductivity was 3 mS/cm.
CEX wash buffer - 100 mM MOPS with 22 mM NaCI made by adding 19.5 g of
MOPS free acid, 1 .5 g of MOPS sodium salt, and 1 .28 g of NaCI per L of buffer. The final pH was 5.7-6.6 (as desired) and the final conductivity was 3 mS/cm. CEX elution buffer - 100 mM MOPS with 1 18 mM NaCI made by adding 19.5 g of MOPS free acid, 1 .5 g of MOPS sodium salt, and 6.93 g of NaCI per L of buffer. The final pH was 5.7-6.6 (as desired) and the final conductivity was 12.4 mS/cm.
CEX strip buffer - 10 mM acetic acid with 3 M NaCI made by adding 0.57 ml_ of glacial acetic acid and 175 g of NaCI per L of buffer. The final pH was 5.3 and the final conductivity was 188 mS/cm.
Lysate preparation. The pH of the lysate was adjusted using 200 mM acetic acid at pH 4.5 made by mixing 1 1 .5 ml_ of glacial acetic acid per L of buffer. The solution was titrated to a final pH of 4.5 using 1 M NaOH. This solution was used in order to avoid localized precipitation due to low pH of concentrated or strong acids.
The load material for these experiments was an extract from a 2 L fed-batch bioreactor run. The extraction of the product from the cell pellet was performed using 20 mM Tris at pH 7.0 with 10 mM EDTA added. The fresh extract was diluted to a conductivity of 3 mS/cm with MilliQ water (typically 1 :1 -1 .5 dilution). The diluted extract was frozen at -20°C in 41 ml_ aliquots. To prepare the load, an aliquot was thawed at room temperature and titrated to pH 5.3 using 200 mM acetic acid at pH 4.5. Small adjustments in conductivity were made by addition of MilliQ water when needed after titration. Finally, the load was diluted 2.5χ to a concentration of ~1 .7 g/L using CEX equilibration buffer. The load was sterile filtered through a 0.8/0.2 μηι filter. The conditioned load was used the same day as prepared.
Host Cell Protein Determination. The host cell protein (HCP) levels were determined by an enzyme-linked immunosorbent assay (ELISA) kit specific for an E. coli expression system (CN F410, Cygnus Technologies). The protocol supplied with the kit was followed exactly. Samples to be assayed for HCP levels were diluted using sample diluent (CN I028, Cygnus Technologies) with a minimum dilution of 10χ. Samples were typically run at two dilutions and plated in duplicate for each dilution. The level of HCP is represented in terms of parts per million (ppm) or ng-HCP/mg-product.
SDS-PAGE Analysis. For purity analysis by sodium dodecyl sulfate
polyacrylamide gel electrophoresis (SDS-PAGE), either 1 mmx 10 well or 1 mmx 15 well NuPAGE 4-12% BisTris gels (CN NP0322Box and NP0323Box, respectively, Invitrogen) are used. The running buffer is 1 χ MES SDS running buffer prepared from a 20χ concentration (CN NP0002, Invitrogen). Novex sharp prestained protein standards (CN 57318, Invitrogen) are used as molecular weight indicators. Samples for analysis were prepared by dilution with MilliQ water to a final volume of 30 μΙ_ and 10 μΙ_ of 4x Lithium dodecyl sulfate (LDS) (CN NP0008, Invitrogen) was added to a final concentration of 1 χ . The samples were mixed by vortex for 5 s. Based on the calculated protein
concentration from the A280/A320 measurement, a target of 3 μg per well was loaded.
wCEX. P05 was evaluated by weak cation exchange chromatography (wCEX) using a Dionex ProPac® WCX-10 4x250 mm column (Product Number 054993), with a flow rate of 1 .2 mL/min using mobile phase solutions of 10 mM sodium acetate pH 5.5 (buffer A) and 10 mM sodium acetate pH 5.5, 250 mM NaCI (buffer B). A gradient is performed from 10%B to 25%B over 20 minutes. Intact P05 elutes approximately 1 .5 to 2.5 minutes before the des-Ala species in the later part of the gradient.
Cation Exchange Capture Chromatography. Chromatography was performed on an AKTA Explorer 100 chromatography system. A 10 ml_ PorosXS column was packed. Material was loaded at 40 mg of P05/ml, or can be loaded at 25-30 mg of P05/ml. The chromatography method is summarized in Table 9. The residence time was held constant during loading and elution steps at 2 min. Mock elution pools were made and assayed for product recovery, HCP level, and % intact protein. A total of 2 runs were completed on the 10 ml_ column with %B at 30% and 35% at pH 6.0.
Table 9
Figure imgf000091_0001
Wash #2 results in a peak comprised mainly of impurities. Elutions #1 with 30% and 35% B result in >95% pure product by SDS-PAGE analysis. The salt concentration of Wash #2 can be increased by a small amount to remove the shoulder on the elution peak.
Example 10
P04 was purified, and diffraction quality crystals were grown in 25% PEG1500, 0.1 M PCB (pH 4.0) at 20QC. The protein crystallized in the space group P212121 , with typical unit cell dimensions of a = 44.5, b = 46.4, c = 64.8. The crystals diffracted to high resolution, and a dataset extending to 1 .47 A was collected at the Advanced Photon Source, beamline LS-CAT 21 ID-F (Chicago IL, USA). The X-ray structure of P04 was solved by molecular replacement using a model incorporating the relevant portions of known IL-1 β and IL-1 Ra structures from PDB structures 1 1TB and 1 1RA (Vigers et al., (1997) Nature 386: 190-194 and Schreuder et al., (1997) Nature 386: 194-200). The final model was refined to a Rwork Rfree of 17.6%/20.4% and contains one P04 molecule (140 residues) and 98 water molecules (Table 10). P04 residues 1 -2, 48-49 and 85-93 were not visible in the electron density and are missing in the final model.
Table 10
Crystallographic Data Collection and Refinement Statistics
Data Collection Statistics
Space group P2i 2i 2i
Cell dimensions
a, b, c (A) 44.5, 46.4, 64.8
α, β, Y ( °) 90, 90, 90
Wavelength (A) 0.9787
Resolution (A) 50.0 - 1 .47 ( 1 .52 - 1 .47)
Emerge 0.039(0.56)
Ι/σΙ 37.7 (2.9)
Completeness (%) 99.8 ( 1 00)
Redundancy 6 (5.9)
Refinement Statistics
Resolution (A) 28.64 - 1 .47 (1 .53 - 1 .47)
No. of reflections 22926
Rwork/Rfree 1 7.6/20.4
Average B (A) 22.3
Rmsd bond lengths (A) 0.007
Rmsd bond angles ( °) 1 .1 37
Numbers in parentheses correspond to highest resolution shell Hnerge = ∑ hkl [∑ i I li " <l> I /∑Ji]
^work = ∑hki II Fobs I - I Fca|C ll/∑hk| I Fobs I where Fobs and Fca,c are the observed and calculated structure factors
Rfree was calculated from a subset of reflections (5%) not used for refinement.
The P04 crystal structure has a fold similar to that predicted by the modeling. A view of this structure is shown in FIG. 1 . An overlay of the two structures is shown in FIG. 12A. The RMSD for backbone carbons was 1 .41 A and 1 .09 A for the IL-1 β and IL-1 Ra segments versus the same residues on the respective parent molecules. P04 has at least one unique salt bridge and two unique hydrogen bonds that involve an interaction between a residue derived from IL-1 β and a counterpart derived from IL-1 Ra: (i) Glu39- Lys64 (Glu39 from IL-1 RA; Lys64 from IL-1 β) as shown in FIG. 12B, (ii) Arg9-Gln149 (Arg9 from IL-1 RA; Gln149 from IL-1 β) as shown in FIG. 12C and (iii) Ser152-Lys40 (Ser152 from IL-1 RA; Lys50 from IL-1 β) as shown in FIG. 12C. These unique
interactions can explain P04's increased thermal stability as these interactions are absent in the I L- 1 β and IL-1 Ra structures. The residues involved in these interactions are also present in P03 and P05, proteins that likewise have increased thermal stability.
Other embodiments are within the scope of the following claims.

Claims

What is claimed is:
1 . An isolated protein that comprises a chimeric interleukin-1 (IL-1 ) family cytokine domain wherein at least a first segment of the domain is at least 20 amino acids in length and has at least 80% amino acid identity to a corresponding segment of a first IL-1 family cytokine, and
at least a second segment of the domain is at least 20 amino acids in length and has at least 80% amino acid identity to a corresponding segment of a second IL-1 family cytokine.
2. The protein of claim 1 wherein the first and second IL-1 family cytokines are selected from the group consisting of IL-1 β, IL-1 a, and IL-1 Ra.
3. The protein of claim 2 wherein the first IL-1 family cytokine is IL-1 β and the second IL-1 family cytokine is IL-1 Ra.
4. The protein of claim 2 wherein the protein binds to IL1 receptor I and inhibits IL-1 receptor I activity.
5. The protein of claim 2 wherein the first segment of the cytokine domain has length at least 20 or 30 amino acids and is identical to a corresponding segment of IL-1 β.
6. The protein of claim 2 wherein the cytokine domain comprises Site A residues that are at least 80% identical to corresponding residues in IL-1 Ra and Site B residues that are at least 80% identical to corresponding residues in IL-1 β .
7. The protein of claim 3 wherein the cytokine domain includes (a) amino acids identical to IL-1 Ra at the following positions ARG1 1 , SER13, GLN14, GLN15, GLU25, LYS27, LEU29, HIS30, LEU31 , GLN32, GLY33, GLN34, ASP35, MET36, GLN38,
GLN39, ALA127, GLU128, ASN129, MET130, and GLN141 (according to the numbering of IL-1 β) and
(b) amino acids identical to IL-1 β at the following positions: ALA1 , PR02, VAL3, ARG4, LEU6, PHE46, GLN48, GLU51 , SER52, ASN53, LYS55, ILE56, PR057, LYS92, LYS93, LYS94, LYS103, GLU105, ASN108, GLN149, PHE150, and SER152 (according to the numbering of IL-1 β).
8. The protein of claim 2 wherein the cytokine domain comprises sequences that are identical to beta strands β2, β3, β10 and β1 1 of IL-1 Ra and sequences that are identical to beta strands β4, β6, β7, and β8 of IL-1 β.
9. The protein of claim 2 wherein one or more of the β1 β2, β2β3, β8β9, β10β1 1 , and β1 1 β12 loops are at least 90% identical to corresponding loops from IL-1 Ra.
10. The protein of claim 2 wherein the cytokine domain comprises at least two discontinuous segments of length at least 20 amino acids that are at least 90% identical to respective corresponding segments from IL-1 β .
1 1 . The protein of claim 10 wherein the two discontinuous segments are identical to respective corresponding segments from IL-1 β .
12. The protein of claim 10 wherein the two discontinuous segments are between 25-40 amino acids in length.
13. The protein of claims 10, 1 1 , or 12, wherein amino acids not in the two discontinuous segments are at least 90% identical to IL-1 Ra.
14. The protein of claim 2 wherein the protein consists of a single polypeptide chain that is 150-160 amino acids in length.
15. The protein of claim 2 wherein the cytokine domain is identical to the first IL-1 family cytokine at at least 50 positions and is identical to the second IL-1 family cytokine at at least 50 positions.
16. The protein of claim 2 wherein at least residues 1 1 -41 and 120-147
(according to IL-1 β numbering) are collectively at least 90% identical to corresponding residues in IL-1 Ra.
17. The protein of claim 3 wherein at least residues 1 -6, 45-61 and 148-153 (according to IL-1 β numbering) are collectively at least 90% identical to corresponding residues in IL-1 β.
18. The protein of claim 3 wherein at least residues 1 -8, 42-120, and 148-153 (according to IL-1 β numbering) are collectively at least 90% identical to corresponding residues in IL-1 β.
19. The protein of claim 3 wherein the cytokine domain is at least 80% identical to P01 , P02, P03, P04, or P05.
20. The protein of claim 19 wherein the cytokine domain is at least 90% identical to P01 , P02, P03, P04, or P05.
21 . The protein of claim 20 wherein the cytokine domain is at least 95% identical to P01 , P02, P03, P04, or P05.
22. The protein of claim 21 wherein the cytokine domain is identical to P01 , P02, or P03.
23. The protein of claim 21 wherein the cytokine domain is identical to P04.
24. The protein of claim 21 wherein the cytokine domain is identical to P05.
25. The protein of claim 2 wherein the cytokine domain has greater
thermostability than IL-1 β and IL-1 Ra.
26. The protein of claim 2 wherein the cytokine domain has a Tm at least 2 degrees greater than IL-1 β and IL-1 Ra.
27. The protein of claim 3 wherein the protein has an IC50 less than 10 ng/ml for inhibition of IL-1 β (at 0.1 ng/ml) in a cellular signaling assay.
28. The protein of claim 3 wherein the protein binds to IL-1 Rl with a higher affinity KD, a slower dissociation rate, and/or a faster association rate than human IL-1 Ra.
29. The protein of claim 3 wherein the cytokine domain comprises one or more of the following:
(a) Site A:
i. Site A residues that are at least 80% identical to corresponding residues in a first parental cytokine domain;
ii. Extended Site A residues that are at least 80% identical to corresponding residues in a first parental cytokine domain;
iii. Site A segments A1 and A2 that are at least 80% identical to corresponding segments of a first parental cytokine domain; or iv. a Site A region that is at least 80% identical to corresponding regions of a first parental cytokine domain;
(b) Site B:
i. Site B residues that are at least 80% identical to corresponding residues in a second parental cytokine domain;
ii. Extended Site B residues that are at least 80% identical to
corresponding residues in a second parental cytokine domain;
iii. Site B segments B1 , B2, and B3 that are at least 80% identical to
corresponding segments of a second parental cytokine domain; or iv. a Site B region that is at least 80% identical to corresponding regions of a second parental cytokine domain.
30. An isolated IL-1 inhibitor comprising an IL-1 family cytokine domain that binds to IL-1 Rl and comprises
(i) a C1 segment that is at least 70% identical to corresponding residues in IL-1 Ra, and
(ii) Site A or Site B residues that are at least 80% identical to corresponding residues in IL-1 β, IL-1 a, or IL-1 Ra, and
wherein the domain is between 40% - 90% identical to IL-1 Ra.
31 . The IL-1 inhibitor of claim 30 wherein the C1 segment is at least 80% identical to corresponding residues in IL-1 Ra.
32. The IL-1 inhibitor of claim 30 wherein the C1 segment is identical to corresponding residues in IL-1 Ra.
33. The IL-1 inhibitor of claim 30 wherein the D2 segment is at least 80% identical to corresponding residues in IL-1 Ra.
34. The IL-1 inhibitor of claim 30 wherein the A1 and A2 segments are collectively at least 80% identical to corresponding residues in IL-1 Ra.
35. The IL-1 inhibitor of claim 30 wherein the B1 , B2, and B3 segments are at least 80% identical to corresponding residues in IL-1 β .
36. The IL-1 inhibitor of claim 30 that inhibits signaling by IL-1 β.
37. The IL-1 inhibitor of claim 30 that binds to IL-1 Rl with an affinity of less than 1 nM.
38. An isolated receptor binding protein that comprises:
(a) amino acids identical to IL-1 Ra at the following positions ARG1 1 , SER13,
GLN14, GLN15, GLU25, LYS27, LEU29, HIS30, LEU31 , GLN32, GLY33, GLN34, ASP35, MET36, GLN38, GLN39, ALA127, GLU128, ASN129, MET130, and GLN141 (according to the numbering of IL-1 β) and
(b) amino acids identical to IL-1 β at the following positions: ALA1 , PR02, VAL3, ARG4, LEU6, PHE46, GLN48, GLU51 , SER52, ASN53, LYS55, ILE56, PR057, LYS92, LYS93, LYS94, LYS103, GLU105, ASN108, GLN149, PHE150, and SER152 (according to the numbering of IL-1 β).
39. The protein of claim 38 that is a chimera of IL-1 β and IL-1 Ra.
40. The protein of claim 38 that comprises Gln48-Asn53 of IL-1 β.
41 . The protein of claim 38 that is at least 50% identical to a cytokine other than IL-1 β.
42. The protein of claim 38 that comprises residues identical to IL-1 β at positions corresponding to Ala1 , Pro2, Val3, Arg4 and Leu6.
43. The protein of claim 38 that comprises at least 15 residues identical to IL-1 β at positions corresponding to 45-61 of IL-1 β (SEQ ID NO:1 ).
44. The protein of claim 38 that comprises at least 8 residues identical to
IL-1 β at positions corresponding to 86-95 of IL-1 β (SEQ ID NO:1 ).
45. The protein of claim 38 that comprises at least 5 residues identical to IL-1 β at positions corresponding to 148-153 of IL-1 β (SEQ ID NO:1 ).
46. The protein of claim 38 wherein the amino acid positions corresponding to residues 1 1 -36 and 126-130 (according to SEQ ID NO:1 ) are predominantly identical to IL-1 Ra.
47. The protein of claim 38 wherein the amino acid positions corresponding to residues 9-41 and 121 -140 (according to SEQ ID NO:1 ) are predominantly identical to IL-1 Ra.
48. A pharmaceutical composition comprising a protein according to any of claims 1 -47.
49. A topical pharmaceutical composition comprising a protein according to any of claims 1 -47.
50. An ophthalmic pharmaceutical composition comprising a protein according to any of claims 1 -47.
51 . The pharmaceutical composition of claim 48 that is aqueous and comprises the protein at a concentration of between 0.001 - 5%.
52. Use of a composition according to claim 48 to a subject to modulate an immune or inflammatory response in a subject.
53. Use of a composition according to claim 48 to treat a subject who has or at risk for a systemic autoimmune disorder.
54. Use of a composition according to claim 48 to treat a subject who has or at risk for rheumatoid arthritis.
55. Use of a composition according to claim 48 to treat a subject having or at risk for dry eye disease or allergic conjunctivitis.
56. An isolated nucleic acid comprising a sequence encoding the protein of any of claims 1 to 47.
57. An isolated nucleic acid vector comprising a sequence encoding the protein of any of claims 1 to 47, wherein the sequence is operably linked to a transcription control sequence.
58. A recombinant host cell comprising a recombinant nucleic acid encoding the protein of any of claims 1 to 47.
59. The host cell of claim 58 that is an E. coli host cell.
60. A method of preparing a recombinant protein, the method comprising culturing the host cell of claim 58 under conditions that permit expression of the recombinant protein;
purifying the recombinant protein from cell lysate or cell media; and
formulating the recombinant protein for administration to a subject.
61 . An isolated protein comprising a chimeric IL-1 family cytokine domain comprising at least two discontinuous segments of length at least six amino acids and having at least 80% identity to corresponding segments of a first IL-1 family cytokine and comprising amino acids predominantly from a second IL-1 family cytokine at remaining positions.
62. The protein of claim 61 , wherein the discontinuous segments comprises residues 1 -6 and 45-61 , 1 -6 and 86-95, 45-61 and 86-95, 1 -6 and 148-153, 45-61 and 148-153, or 86-95 and 148-153, according to the numbering of such positions in IL-1 β.
63. The protein of claim 61 wherein the protein comprises three discontinuous segments having amino acids identical to those in corresponding segments of the first IL 1 family cytokine, and those segments include residues 1 -8, 42-120, and 141 -153.
64. The protein of claim 61 wherein the protein comprises four discontinuous segments having amino acids identical to those in corresponding segments of the first IL 1 family cytokine, and those segments include residues 1 -6, 45-61 , 86-95, and 148-153.
65. The protein of claim 61 wherein the protein comprises three discontinuous segments having amino acids identical to those in corresponding segments of the first IL 1 family cytokine, and those segments include residues 1 -10, 37-125, and 131 -153.
66. The protein of claim 61 wherein the protein is identical to the second IL-1 family cytokine at the remaining positions.
67. The protein of claim 61 wherein one or more of the borders of the discontinuous segments are located at positions wherein the first and second IL-1 family cytokines have the same amino acids.
68. The protein of claim 61 wherein the protein is identical to the first IL-1 family cytokine at at least 50 positions and is identical to the second IL-1 family cytokine at at least 50 positions.
69. The protein of claim 61 wherein the first and second IL-1 family cytokines are selected from the group consisting of IL-1 β, IL-1 a, and IL-1 Ra.
70. The protein of claim 69 wherein the first IL-1 family cytokine is IL-1 β and the second IL-1 family cytokine is IL-1 Ra.
PCT/US2011/045995 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists WO2012016203A1 (en)

Priority Applications (25)

Application Number Priority Date Filing Date Title
EP18191564.6A EP3470421B9 (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i antagonists
BR112013003646-0A BR112013003646B1 (en) 2010-07-29 2011-07-29 ISOLATED PROTEIN THAT COMPRISES A DOMAIN OF CYTOKINE OF THE INTERLEUKIN-1 (IL-1) CHIMERIC FAMILY, ITS METHOD OF PREPARATION, PHARMACEUTICAL COMPOSITION AND USES, AS WELL AS, NUCLEIC ACID, VECTOR, AND HOST CELL
CN201180047508.1A CN103221422B (en) 2010-07-29 2011-07-29 1 receptor type I agonist of chimeric IL and antagonist
EP23157789.1A EP4219538A3 (en) 2010-07-29 2011-07-29 Receptor binding agents
SG2013006697A SG187231A1 (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists
EP11749612.5A EP2598526B1 (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists
US13/812,582 US8853150B2 (en) 2010-07-29 2011-07-29 Chimeric IL-1 receptor type I antagonists
DK11749612.5T DK2598526T3 (en) 2010-07-29 2011-07-29 CHEMICAL IL-1 RECEPTOR TYPE-I AGONISTS AND ANTAGONISTS
RU2013108836/10A RU2013108836A (en) 2010-07-29 2011-07-29 TYPE I-1 HYBRID RECEPTOR AGONISTS AND ANTAGONISTS
KR1020137005337A KR102080704B1 (en) 2010-07-29 2011-07-29 Chimeric IL-1 Receptor Type 1 Agonists and Antagonists
PL11749612T PL2598526T3 (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists
JP2013522011A JP6030554B2 (en) 2010-07-29 2011-07-29 Chimeric IL-1 receptor type 1 agonists and antagonists
NZ605928A NZ605928A (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists
SI201131571T SI2598526T1 (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists
ES11749612T ES2701405T3 (en) 2010-07-29 2011-07-29 Agonists and antagonists of the type I receptor of chimeric IL-1
LTEP11749612.5T LT2598526T (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists
CA2806438A CA2806438C (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists
AU2011283669A AU2011283669A1 (en) 2010-07-29 2011-07-29 Chimeric IL-1 receptor type I agonists and antagonists
MX2013001107A MX360097B (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists.
IL224435A IL224435B (en) 2010-07-29 2013-01-28 Chimeric il-1 receptor type i agonists and antagonists
US14/460,563 US9458216B2 (en) 2010-07-29 2014-08-15 Nucleic acid encoding chimeric IL-1 receptor type I antagonists
AU2016213845A AU2016213845B2 (en) 2010-07-29 2016-08-12 Chimeric il-1 receptor type i agonists and antagonists
IL259547A IL259547B (en) 2010-07-29 2018-05-23 Isolatrd protein and composition comprising same for use for the treatment of il-1 mediated disorders
HRP20181446TT HRP20181446T1 (en) 2010-07-29 2018-09-10 Chimeric il-1 receptor type i agonists and antagonists
AU2018250431A AU2018250431A1 (en) 2010-07-29 2018-10-18 Chimeric il-1 receptor type i agonists and antagonists

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
US36879910P 2010-07-29 2010-07-29
US61/368,799 2010-07-29
US201161436178P 2011-01-25 2011-01-25
US201161436184P 2011-01-25 2011-01-25
US61/436,184 2011-01-25
US61/436,178 2011-01-25
US201161493967P 2011-06-06 2011-06-06
US201161493966P 2011-06-06 2011-06-06
US61/493,966 2011-06-06
US61/493,967 2011-06-06

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/812,582 A-371-Of-International US8853150B2 (en) 2010-07-29 2011-07-29 Chimeric IL-1 receptor type I antagonists
US14/460,563 Division US9458216B2 (en) 2010-07-29 2014-08-15 Nucleic acid encoding chimeric IL-1 receptor type I antagonists

Publications (1)

Publication Number Publication Date
WO2012016203A1 true WO2012016203A1 (en) 2012-02-02

Family

ID=44533130

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/045995 WO2012016203A1 (en) 2010-07-29 2011-07-29 Chimeric il-1 receptor type i agonists and antagonists

Country Status (23)

Country Link
US (2) US8853150B2 (en)
EP (3) EP4219538A3 (en)
JP (1) JP6030554B2 (en)
KR (1) KR102080704B1 (en)
CN (2) CN107337735B (en)
AU (3) AU2011283669A1 (en)
BR (1) BR112013003646B1 (en)
CA (1) CA2806438C (en)
DK (1) DK2598526T3 (en)
ES (2) ES2701405T3 (en)
HK (1) HK1246327A1 (en)
HR (2) HRP20230776T1 (en)
HU (2) HUE063115T2 (en)
IL (2) IL224435B (en)
LT (1) LT2598526T (en)
MX (1) MX360097B (en)
NZ (1) NZ605928A (en)
PL (2) PL3470421T3 (en)
RU (2) RU2013108836A (en)
SG (1) SG187231A1 (en)
SI (1) SI2598526T1 (en)
WO (1) WO2012016203A1 (en)
ZA (1) ZA201505145B (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012103240A3 (en) * 2011-01-25 2012-10-26 Eleven Biotherapeutics, Inc. Chimeric il-1 receptor type i agonists and antagonists
WO2013019652A1 (en) * 2011-07-29 2013-02-07 Eleven Biotherapeutics, Inc. Purified proteins
WO2014160371A1 (en) 2013-03-13 2014-10-02 Eleven Biotherapeutics, Inc. Chimeric cytokine formulations for ocular delivery
JP2015525217A (en) * 2012-06-08 2015-09-03 アルカーメス,インコーポレイテッド Ligands modified by circulating substitution as agonists and antagonists
WO2015138844A1 (en) 2014-03-13 2015-09-17 Eleven Biotherapeutics, Inc. Methods for production of stable chimeric cytokine protein formulations in blow fill seal containers
US9163072B2 (en) 2007-05-01 2015-10-20 Alligator Bioscience Ab Mutants of interleukin-1 receptor antagonist
US9458216B2 (en) 2010-07-29 2016-10-04 Eleven Biotherapeutics, Inc. Nucleic acid encoding chimeric IL-1 receptor type I antagonists
EP3004154A4 (en) * 2013-06-05 2017-01-04 Ben Gurion University of The Negev Research and Development Authority IL-1beta-IL-1Ra CHIMERA AND METHODS OF USING THE SAME
AU2015268652B2 (en) * 2012-06-08 2017-11-23 Alkermes Pharma Ireland Limited Ligands modified by circular permutation as agonists and antagonists
WO2023169686A1 (en) 2022-03-10 2023-09-14 Buzzard Pharmaceuticals AB Inhibitor of interleukin-1 receptor type 1 for use in the treatment of cancer

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3003356B1 (en) * 2013-05-31 2019-07-03 Regeneron Pharmaceuticals, Inc. Il-1 antagonists for use in treating alzheimer's disease
WO2015048526A1 (en) * 2013-09-27 2015-04-02 Eleven Biotherapeutics, Inc. Treating ocular pain
EP3871682A1 (en) * 2016-10-14 2021-09-01 i.com Medical GmbH Method for establishing, restoring, and preserving homeostasis of the ocular surface
CN114480406B (en) * 2021-09-16 2024-01-30 广东翠点生物科技有限公司 IL-1 signal path response element and application thereof

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0036776A2 (en) 1980-03-24 1981-09-30 Genentech, Inc. A method of creating an expression plasmid
US4804539A (en) 1986-07-28 1989-02-14 Liposome Technology, Inc. Ophthalmic liposomes
US4883658A (en) 1986-04-28 1989-11-28 Holly Frank J Ophthalmic solution for treatment of dry-eye syndrome
EP0362179A2 (en) 1988-08-25 1990-04-04 Smithkline Beecham Corporation Recombinant saccharomyces
US5075104A (en) 1989-03-31 1991-12-24 Alcon Laboratories, Inc. Ophthalmic carboxy vinyl polymer gel for dry eye syndrome
US5278151A (en) 1987-04-02 1994-01-11 Ocular Research Of Boston, Inc. Dry eye treatment solution
US5578586A (en) 1987-04-02 1996-11-26 Ocular Research Of Boston, Inc. Dry eye treatment process and solution
US5693489A (en) 1984-03-30 1997-12-02 Associated Universities, Inc. Cloning and expression of the gene for bacteriophage T7 RNA polymerase
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
WO1998047921A1 (en) * 1997-04-21 1998-10-29 Schering Corporation Mammalian cytokines; related reagents and methods
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
WO2002060919A2 (en) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules with extended half-lives, compositions and uses thereof
WO2008155134A1 (en) 2007-06-21 2008-12-24 Technische Universität München Biological active proteins having increased in vivo and/or vitro stability
WO2009023270A2 (en) 2007-08-15 2009-02-19 Amunix, Inc. Compositions and methods for modifying properties of biologically active polypeptides

Family Cites Families (107)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL188266C (en) 1975-07-29 1992-05-18 Merck & Co Inc PROCESS FOR THE PREPARATION OF AN ORGANIC IMPLANT.
US5998578A (en) 1984-05-18 1999-12-07 New England Medical Center Hospitals, Inc. Biologically active fragments of IL-1β
EP0569687B1 (en) 1984-05-18 2002-08-21 New England Medical Center Hospitals, Inc. Human IL-1 cDNA sequences encoding biologically-active human IL-1 proteins
US5484887A (en) 1984-06-19 1996-01-16 Immunex Corporation Homogeneous interleukin 1
US6107465A (en) 1984-12-21 2000-08-22 Otsuka Pharmaceutical Co., Ltd. IL-1β and derivatives thereof and drugs
DK172052B1 (en) 1984-12-21 1997-09-29 Otsuka Pharma Co Ltd Antitumor active substance, method for its preparation, drug containing the substance, gene encoding the substance, vector containing the gene as well as recombinant microorganism transformed with such a vector
US5122459A (en) 1984-12-31 1992-06-16 Immunex Corporation Gene encoding biologically active human interleukin 1
US4770781A (en) 1986-03-03 1988-09-13 Merck & Co., Inc. Purification of human interleukin-1 species
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
US6159460A (en) 1988-05-27 2000-12-12 Amgen Inc. Method for treating interleukin-1 mediated diseases
US5075222A (en) 1988-05-27 1991-12-24 Synergen, Inc. Interleukin-1 inhibitors
KR0148009B1 (en) 1988-05-27 1998-08-01 그래고리 비. 아보트 Interleukin-1 inhibitors
US6858409B1 (en) 1988-05-27 2005-02-22 Amgen Inc. Nucleic acids encoding interleukin-1 inhibitors and processes for preparing interleukin-1 inhibitors
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
DK0460052T3 (en) 1989-02-27 1999-03-08 New England Medical Center Inc Inhibitors of the biological activity of IL-1
EP0502956B1 (en) 1989-11-29 1997-04-23 Amgen Boulder Inc. Production of recombinant human interleukin-1 inhibitor
WO1991017184A1 (en) 1990-04-27 1991-11-14 The Upjohn Company Modified interleukin-1 inhibitors
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
EP0570462A4 (en) 1991-02-05 1994-11-23 Univ Maryland MODIFIED INTERLEUKIN-1-g(b).
DE69233254T2 (en) 1991-06-14 2004-09-16 Genentech, Inc., South San Francisco Humanized Heregulin antibody
DE69230862T2 (en) 1991-10-15 2000-12-14 Michael F Mullarkey Receptors for the treatment of delayed-type inflammation
JP3623501B2 (en) 1991-10-31 2005-02-23 ザ ビクトリア ユニバーシティ オブ マンチェスター Treatment of neurological conditions with interleukin-1 inhibitory compounds
PL175705B1 (en) 1992-09-17 1999-01-29 Synergen Inc Pharmaceutical agents containing interleukin-1 inhibitors
AU688751B2 (en) 1993-01-12 1998-03-19 Biogen Idec Ma Inc. Recombinant anti-VLA4 antibody molecules
US5340572A (en) 1993-02-08 1994-08-23 Insite Vision Incorporated Alkaline ophthalmic suspensions
WO1995010298A1 (en) 1993-10-12 1995-04-20 Mary Lake Polan Method of contraception
US5861476A (en) 1994-02-02 1999-01-19 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
IT1269989B (en) 1994-09-21 1997-04-16 Dompe Spa IL-1 RECEPTOR ANTAGONISTS WITH INCREASED INHIBITORY ACTIVITY
WO1996034096A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6096728A (en) 1996-02-09 2000-08-01 Amgen Inc. Composition and method for treating inflammatory diseases
US6416753B1 (en) 1996-03-15 2002-07-09 The General Hospital Corporation Method for modulating apoptosis
US6974682B1 (en) 1996-08-26 2005-12-13 Human Genome Sciences, Inc. Soluble interleukin-1 receptor accessory molecule
ZA9710414B (en) * 1996-11-19 1999-05-19 Schepens Eye Res Inst Local use of IL-1RA for prophylaxis and treatment of corneal transplant rejection and other immune and inflammatory disorders of the eye
WO1998022130A1 (en) 1996-11-19 1998-05-28 The Schepens Eye Research Institute, Inc. Local use of il-1ra in corneal transplant rejection or disorders of the eye
ES2301183T3 (en) 1996-12-03 2008-06-16 Amgen Fremont Inc. COMPLETELY HUMAN ANTIBODY THAT JOINS THE EGFR RECEIVER.
EP2002846B1 (en) 1996-12-06 2017-01-25 Amgen Inc. Combination therapy using an IL-1 inhibitor for treating IL-1 mediated diseases
US6337072B1 (en) 1998-04-03 2002-01-08 Hyseq, Inc. Interleukin-1 receptor antagonist and recombinant production thereof
US7906481B2 (en) 1998-09-25 2011-03-15 Sciaticon Ab Specific TNF-A inhibitors for treating spinal disorders mediated by nucleous pulposus
US6927044B2 (en) 1998-09-25 2005-08-09 Regeneron Pharmaceuticals, Inc. IL-1 receptor based cytokine traps
US6471961B1 (en) 1999-02-24 2002-10-29 Edward L. Tobinick Interleukin antagonists for the treatment of neurological, retinal and muscular disorders
US6419944B2 (en) 1999-02-24 2002-07-16 Edward L. Tobinick Cytokine antagonists for the treatment of localized disorders
JP3363832B2 (en) 1999-05-13 2003-01-08 アルプス電気株式会社 Thin film structure and method of manufacturing the same
WO2001041792A1 (en) 1999-12-10 2001-06-14 Amgen, Inc. Interleukin-1 receptor antagonist-like molecules and uses thereof
KR20020073139A (en) 1999-12-10 2002-09-19 암젠 인코포레이티드 Interleukin-1 receptor antagonist-like molecules and uses thereof
CA2395417A1 (en) 1999-12-10 2001-06-14 Amgen Inc. Interleukin-1 receptor antagonist-related molecules and uses thereof
US20030007971A1 (en) 2000-01-31 2003-01-09 Hideaki Hara Remedies for ophthalmic diseases
AU2001236632A1 (en) 2000-02-03 2001-08-14 Regeneration Technologies, Inc. Extraction of growth factors from tissue
US20040026011A1 (en) 2000-04-17 2004-02-12 Edwards Mark Stephen Method and apparatus for making bristle subassemblies
US6623736B2 (en) 2000-05-02 2003-09-23 Edward L. Tobinick Interleukin antagonists for the treatment of neurological, retinal and muscular disorders
EP1282435A2 (en) 2000-05-12 2003-02-12 Immunex Corporation Interleukin-1 inhibitors in the treatment of diseases
US20030026806A1 (en) 2000-10-27 2003-02-06 Amgen Inc. Antibodies and other selective IL-1 binding agents that allow binding to IL-1 receptor but not activation thereof
US7087224B2 (en) 2000-10-31 2006-08-08 Amgen Inc. Method of treating anemia by administering IL-1ra
US20030166069A1 (en) 2000-11-28 2003-09-04 Amgen, Inc. Interleukin-1 receptor antagonist-like molecules and uses thereof
KR20030074790A (en) 2001-02-06 2003-09-19 메르크 파텐트 게엠베하 Modified interleukin-1 receptor antagonist (il-1ra) with reduced immunogenicity
US20050143333A1 (en) 2001-05-18 2005-06-30 Sirna Therapeutics, Inc. RNA interference mediated inhibition of interleukin and interleukin receptor gene expression using short interfering nucleic acid (SINA)
US20030083301A1 (en) 2001-09-06 2003-05-01 Board Of Regents, The University Of Texas System Therapeutic treatments for spinal cord injury via blockade of interleukin-1 receptor
AU2003217386B2 (en) 2002-02-13 2008-06-26 Ludwig Institute For Cancer Research Humanized GM-CSF antibodies
AU2003226428A1 (en) 2002-04-18 2003-11-03 Northwestern University Encapsulation of nanotubes via self-assembled nanostructures
US7029712B1 (en) 2002-07-17 2006-04-18 Biosyntrx Inc Treatment for dry eye syndrome
WO2004060911A2 (en) 2002-12-30 2004-07-22 Amgen Inc. Combination therapy with co-stimulatory factors
US20050033694A1 (en) 2003-06-03 2005-02-10 United States Postal Service. System and method for fleet card management
US7261882B2 (en) 2003-06-23 2007-08-28 Reagents Of The University Of Colorado Methods for treating neuropathic pain by administering IL-10 polypeptides
US20050023872A1 (en) 2003-07-28 2005-02-03 Hetzel Thomas R. Modular seat cushion with interlocking human support and base portions and method of creating and using a seat cushion
KR100543466B1 (en) 2003-11-13 2006-01-20 삼성전자주식회사 Image interpolation apparatus and method
US7482323B2 (en) 2004-03-04 2009-01-27 The University Of Tennessee Research Foundation Intracellular interleukin-1 receptor antagonist and uses thereof
US7674464B2 (en) 2004-03-04 2010-03-09 The University Of Tennessee Research Foundation Intracellular interleukin-1 receptor antagonists
ES2694252T3 (en) 2004-04-02 2018-12-19 Swedish Orphan Biovitrum Ab (Publ) Methods to reduce the aggregation of IL-1ra
US8618054B2 (en) 2004-05-05 2013-12-31 Valorisation-Rechereche Société en Commandite Interleukin-1 receptor antagonists, compositions, and methods of treatment
WO2005117945A1 (en) 2004-06-04 2005-12-15 Regeneron Pharmaceuticals, Inc. Methods of using il-1 antagonists to treat autoinflammatory disease
PL1778723T3 (en) 2004-08-17 2013-03-29 Regeneron Pharma Il-1 antagonist formulations
EP1781684A4 (en) * 2004-08-25 2008-05-21 Amprotein Corp A novel chimeric polypeptide and use thereof
US20060110429A1 (en) 2004-11-24 2006-05-25 Therakine Corporation Implant for intraocular drug delivery
RU2007119989A (en) 2004-12-02 2009-01-10 Домантис Лимитед (Gb) COMPOSITIONS, FUSED CONSTRUCTIONS AND PLAD DOMAIN CONJUGATES
WO2007014073A2 (en) 2005-07-22 2007-02-01 Amgen Inc. Concentrated protein lyophilates, methods, and uses
CA2621086A1 (en) 2005-09-01 2007-03-08 Schering Corporation Use of il-23 and il-17 antagonists to treat autoimmune ocular inflammatory disease
IL172297A (en) 2005-10-03 2016-03-31 Compugen Ltd Soluble vegfr-1 variants for diagnosis of preeclamsia
EP1948220A2 (en) 2005-10-21 2008-07-30 Amgen Inc. Methods of decreasing vascular calcification using il-1 inhibitors
BRPI0709977A2 (en) 2006-04-14 2011-08-02 Novartis Ag Use of IL-1 Antibodies for the Treatment of Ophthalmic Disorders
WO2007123976A2 (en) 2006-04-18 2007-11-01 The Board Of Trustees Of The Leland Stanford Junior University Antibody profiling for determination of patient responsiveness
WO2007145618A1 (en) 2006-06-12 2007-12-21 Therakine Limited Topical treatment for diseases of eye surface
US20080095754A1 (en) 2006-10-18 2008-04-24 Burke Susan E Ophthalmic compositions comprising diglycine
US8399421B2 (en) 2007-03-30 2013-03-19 The Board Of Regents Of The University Of Texas System Treatment for neuropathic pain due to spinal cord injury
GB0708376D0 (en) 2007-05-01 2007-06-06 Alligator Bioscience Ab Novel polypeptides and uses thereof
EP2468302A1 (en) 2007-05-29 2012-06-27 Novartis AG New indications for anti-IL-1beta therapy
KR101430584B1 (en) 2007-07-05 2014-08-14 삼성전자주식회사 Apparatus and method for providing sim application toolkit in mobile communication system
JP5894364B2 (en) 2007-08-16 2016-03-30 ザ スキーペンズ アイ リサーチ インスティチュート インコーポレイテッド Therapeutic composition for treating inflammation of eye and appendage tissue
AU2008310902A1 (en) 2007-10-08 2009-04-16 Anaphore, Inc. Trimeric IL-1Ra
JP5886523B2 (en) 2008-01-09 2016-03-16 ザ スキーペンズ アイ リサーチ インスティチュート インコーポレイテッド Therapeutic composition for treating inflammatory disorders of the eye
WO2010089522A1 (en) 2009-07-16 2010-08-12 Arecor Limited Stable formulation of a therapeutic protein
US8298533B2 (en) 2008-11-07 2012-10-30 Medimmune Limited Antibodies to IL-1R1
EP2385839B1 (en) 2009-01-09 2014-11-26 The Schepens Eye Research Institute, Inc. Therapeutic compositions for treatment of corneal disorders
US20120014970A1 (en) 2009-01-09 2012-01-19 Reza Dana Therapeutic Compositions for Treatment of Corneal Disorders
CN102395378B (en) 2009-02-18 2016-01-20 爱昂粒子科学有限责任公司 For the bifunctional copolymer of ophthalmology and other externals or topical application
EP2248518B1 (en) 2009-04-17 2013-01-16 Merz Pharma GmbH & Co. KGaA Formulation for stabilizing proteins, peptides or mixtures thereof.
US20120269765A1 (en) 2009-07-24 2012-10-25 Garcia K Christopher Cytokine compositions and methods of use thereof
WO2011028344A2 (en) 2009-08-25 2011-03-10 Amunix Operating Inc. Interleukin-1 receptor antagonist compositions and methods of making and using same
WO2011044563A2 (en) 2009-10-10 2011-04-14 The Board Of Trustees Of The Leland Stanford Junior University Il-17 family cytokine compositions and uses
WO2011063195A2 (en) 2009-11-20 2011-05-26 Dr. Reddy's Laboratories Ltd. Purification of modified cytokines
WO2011106697A1 (en) 2010-02-25 2011-09-01 Schepens Eye Research Institute Therapeutic compositions for the treatment of dry eye disease
WO2011123813A2 (en) 2010-04-02 2011-10-06 Amunix Operating Inc. Binding fusion proteins, binding fusion protein-drug conjugates, xten-drug conjugates and methods of making and using same
US20130195868A1 (en) 2010-06-24 2013-08-01 Eleven Biotherapeutics, Inc. Treating surface of the eye disorders
CA2806438C (en) 2010-07-29 2021-04-13 Eleven Biotherapeutics, Inc. Chimeric il-1 receptor type i agonists and antagonists
WO2012103240A2 (en) 2011-01-25 2012-08-02 Eleven Biotherapeutics, Inc. Receptor binding agents
ES2600616T3 (en) 2011-03-14 2017-02-10 Phlogo Aps Interleukin-1 receptor antagonists
WO2013019652A1 (en) 2011-07-29 2013-02-07 Eleven Biotherapeutics, Inc. Purified proteins
JP6450364B2 (en) 2013-03-13 2019-01-09 セセン バイオ, インコーポレイテッド Chimeric cytokine formulations for ocular delivery

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0036776A2 (en) 1980-03-24 1981-09-30 Genentech, Inc. A method of creating an expression plasmid
US5693489A (en) 1984-03-30 1997-12-02 Associated Universities, Inc. Cloning and expression of the gene for bacteriophage T7 RNA polymerase
US4883658A (en) 1986-04-28 1989-11-28 Holly Frank J Ophthalmic solution for treatment of dry-eye syndrome
US4804539A (en) 1986-07-28 1989-02-14 Liposome Technology, Inc. Ophthalmic liposomes
US5578586A (en) 1987-04-02 1996-11-26 Ocular Research Of Boston, Inc. Dry eye treatment process and solution
US5278151A (en) 1987-04-02 1994-01-11 Ocular Research Of Boston, Inc. Dry eye treatment solution
EP0362179A2 (en) 1988-08-25 1990-04-04 Smithkline Beecham Corporation Recombinant saccharomyces
US5075104A (en) 1989-03-31 1991-12-24 Alcon Laboratories, Inc. Ophthalmic carboxy vinyl polymer gel for dry eye syndrome
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
WO1998047921A1 (en) * 1997-04-21 1998-10-29 Schering Corporation Mammalian cytokines; related reagents and methods
WO2000042072A2 (en) 1999-01-15 2000-07-20 Genentech, Inc. Polypeptide variants with altered effector function
WO2002060919A2 (en) 2000-12-12 2002-08-08 Medimmune, Inc. Molecules with extended half-lives, compositions and uses thereof
WO2008155134A1 (en) 2007-06-21 2008-12-24 Technische Universität München Biological active proteins having increased in vivo and/or vitro stability
WO2009023270A2 (en) 2007-08-15 2009-02-19 Amunix, Inc. Compositions and methods for modifying properties of biologically active polypeptides

Non-Patent Citations (135)

* Cited by examiner, † Cited by third party
Title
"Current Protocols in Immunology", 1995, JOHN WILEY & SONS, INC
"Current Protocols in Immunology", JOHN WILEY & SONS, INC
"Current Protocols in Molecular Biology", 1989, GREENE PUBLISHING ASSOCIATES
"Current Protocols in Molecular Biology", 1989, JOHN WILEY & SONS
"Handbook of Pharmaceutical Excipients", 2000
"In Vitro Mutagenesis Protocols", 2010, HUMANA PRESS
"Molecular Cloning: A Laboratory Manual", 2001, COLD SPRING HARBOR PRESS
"PCR Cloning Protocols: From Molecular Cloning to Genetic Engineering", 2002, HUMANA PRESS
"Protein formulation and delivery", 2007, INFORMA HEALTH CARE
"Sustained and Controlled Release Drug Delivery Systems", 1978, MARCEL DEKKER, INC.
A WINGREN: "Fusion of a Signal Sequence to the Interleukin-1[beta] Gene Directs the Protein from Cytoplasmic Accumulation to Extracellular Release", CELLULAR IMMUNOLOGY, vol. 169, no. 2, 1 May 1996 (1996-05-01), pages 226 - 237, XP055014943, ISSN: 0008-8749, DOI: 10.1006/cimm.1996.0113 *
AKSENTIJEVICH ET AL., ARTHRITIS RHEUM, vol. 46, 2002, pages 3340 - 3348
ALT, M., J. HEPATOL., vol. 23, 1995, pages 746 - 58
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
ALTSCHUL ET AL., METHODS IN ENZYMOLOGY, vol. 266, 1996, pages 460 - 480
ANSEL ET AL.: "Pharmaceutical Dosage Forms and Drug Delivery Systems", 1999, LIPPINCOTT WILLIAMS & WILKINS
BAKER, SALI, SCIENCE, vol. 294, no. 5540, 2001, pages 93 - 6
BARBINO ET AL., INVEST. OPHTHAL. VIS. SCI., vol. 46, 2005, pages 2766 - 2711
BARDWELL ET AL., RHEUMATOLOGY, vol. 41, no. 1, 2002, pages 38 - 45
BERGE, S.M. ET AL., J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
BEYER ET AL., J. MOL. BIOL., 2008
BODER ET AL., METHODS ENZYMOL., vol. 328, 2000, pages 430 - 44
BOLTON, C., MULTIPLE SCLEROSIS, vol. 143, 1995
BORASCHI ET AL., FRONTIERS IN BIOSCIENCE: A JOURNAL AND VIRTUAL LIBRARY, vol. 1, 1996, pages D270 - 308
BORASCHI, J. IMMUNOL., vol. 155, no. 10, 1995, pages 4719 - 25
BRODY, S. L., CRYSTAL, R. G., ANN. N.Y. ACAD. SCI., vol. 716, 1994, pages 90 - 101
CARTER ET AL., NUCL. ACIDS RES., vol. 13, 1986, pages 4331
CASE ET AL., J. COMPUTAT. CHEM., vol. 26, 2005, pages 1668 - 1688
CASE ET AL.: "AMBER", vol. 11, 2010, UNIVERSITY OF CALIFORNIA
CHANG ET AL., NATURE, vol. 275, 1978, pages 615
CHAO ET AL., NAT PROTOC., vol. 1, no. 2, 2006, pages 755 - 68
CHARLES A. DINARELLO: "Immunological and Inflammatory Functions of the Interleukin-1 Family", ANNUAL REVIEW OF IMMUNOLOGY, vol. 27, no. 1, 1 April 2009 (2009-04-01), pages 519 - 550, XP055014922, ISSN: 0732-0582, DOI: 10.1146/annurev.immunol.021908.132612 *
CHOTHIA, J. MOL. BIOL., vol. 150, 1976, pages 1
COHEN, S. ET AL., ARTHRITIS & RHEUMATISM, vol. 46, 2002, pages 614 - 24
COLBY ET AL., METHODS ENZYMOL., vol. 388, 2004, pages 348 - 58
CREIGHTON: "The Proteins", W. H. FREEMAN & CO.
CUNNINGHAM, WELLS, SCIENCE, vol. 244, 1989, pages 1081 - 1085
DANA ET AL., TRANSPLANTATION, vol. 63, 1997, pages 1501 - 7
DANA, TRANS AM OPHTHALMOL SOC, vol. 105, 2007, pages 330 - 43
DEBOER ET AL., PROC. NATL. ACAD. SCI. USA, vol. 80, 1983, pages 21 - 25
DEH-MING CHANG ET AL: "Dual biological functions of an interleukin-1 receptor antagonist-interleukin-10 fusion protein and its suppressive effects on joint inflammation", IMMUNOLOGY, vol. 112, no. 4, 1 August 2004 (2004-08-01), pages 643 - 650, XP055014940, ISSN: 0019-2805, DOI: 10.1111/j.1365-2567.2004.01910.x *
DEKARIS ET AL., CURR EYE RES, vol. 19, no. 5, 1999, pages 456 - 9
DEMIRCAN ET AL., EYE, vol. 20, 2006, pages 1366 - 1369
DENNIS ET AL., J. BIOL. CHEM., vol. 277, 2002, pages 35035 - 35043
DEUTSCHER, METHODS IN ENZYMOLOGY, vol. 182, 1990
DINARELLO ET AL.: "Current Protocols in Immunology", 2000, JOHN WILEY AND SONS INC.
DOGANAY ET AL., EYE, vol. 16, 2006, pages 163 - 170
DUNCAN ET AL., MOLEC. MED. TODAY, 1997, pages 554 - 561
ECONOMIDES ET AL., NATURE MED., vol. 9, 2003, pages 47 - 52
ENRIQUEZ DE SALAMANCA ET AL., MOL. VIS., vol. 16, 2010, pages 862 - 873
EVANS ET AL., J. BIOL. CHEM., vol. 270, 1995, pages 11477
FELDMANN ET AL., AM J HUM GENET, vol. 71, 2002, pages 198 - 203
FERRY, N., HEARD, J. M., HUM. GENE THER., vol. 9, 1998, pages 1975 - 81
FINZEL ET AL., J. MOL. BIOL., vol. 209, 1989, pages 779 - 791
FLEISCHMANN, R.M. ET AL., ARTHRITIS & RHEUMATISM, vol. 48, 2003, pages 927 - 34
GENNARO: "Remington: The Science and Practice of Pharmacy", 2000, LIPPINCOTT, WILLIAMS & WILKINS
GHETIE ET AL., ANN. REV. IMMUNOL., vol. 18, 2000, pages 739 - 766
GLUZMAN ET AL., CELL, vol. 23, 1981, pages 175
GOEDDEL ET AL., NATURE, vol. 281, 1979, pages 544
GOEDDEL, NUCLEIC ACIDS RES., vol. 8, 1980, pages 4057
GREENFEDER ET AL., J. BIOL. CHEM., vol. 270, 1995, pages 22460
HAL M. HOFFMAN ET AL: "Efficacy and safety of rilonacept (interleukin-1 trap) in patients with cryopyrin-associated periodic syndromes: Results from two sequential placebo-controlled studies", ARTHRITIS & RHEUMATISM, vol. 58, no. 8, 30 July 2008 (2008-07-30), pages 2443 - 2452, XP055014926, ISSN: 0004-3591, DOI: 10.1002/art.23687 *
HANES ET AL., METHODS ENZYMOL., vol. 328, 2000, pages 404 - 30
HANES ET AL., NAT BIOTECHNOL., vol. 18, 2000, pages 1287 - 92
HASSAN ET AL., PLOS ONE., vol. 5, no. 1, 25 January 2010 (2010-01-25), pages E8868
HE ET AL., J. PHARM. SCIENCES, vol. 99, 2010, pages 1707 - 1720
HEIDARY ET AL: "Long-range Coupling Between Separate Docking Sites in Interleukin-1beta", JOURNAL OF MOLECULAR BIOLOGY, ACADEMIC PRESS, UNITED KINGDOM, vol. 353, no. 5, 11 November 2005 (2005-11-11), pages 1187 - 1198, XP005152798, ISSN: 0022-2836, DOI: 10.1016/J.JMB.2005.08.072 *
HESS ET AL., J. ADV. ENZYME REG., vol. 7, 1968, pages 149
HIGGIN ET AL., METHODS ENZYMOL., vol. 266, 1996, pages 383 - 402
HINTON, J. BIOL. CHEM., vol. 279, 2004, pages 6213 - 6216
HIRANO ET AL., J PHARMACOL SCI., vol. 110, no. 2, June 2009 (2009-06-01), pages 169 - 81
HITZEMAN ET AL., J. BIOL. CHEM., vol. 255, 1980, pages 2073
HOFFMAN ET AL., NAURE, vol. 29, 2001, pages 301 - 305
HOLLAND, BIOCHEMISTRY, vol. 17, 1978, pages 4900
IMREN ET AL., PNAS, vol. 99, 2002, pages 14380
INABA ET AL., INFLAMM BOWEL DIS., 2010
ISSEKUTZ, A. C. ET AL., IMMUNOLOGY, vol. 88, 1996, pages 569
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, PUBLIC HEALTH SERVICE
KARLIN ET AL., PNAS USA, vol. 90, 1993, pages 5873 - 5787
KAUFMAN, R. J., LARGE SCALE MAMMALIAN CELL CULTURE, 1990, pages 1569
KAY, M. A., CHEST, vol. 111, no. 6, 1997, pages 138S - 142S
KEANE-MYERS AM ET AL., INVEST OPHTHALMOL VIS SCI, vol. 40, no. 12, 1999, pages 3041 - 6
KRELIN ET AL., CANCER RES., vol. 67, 2007, pages 1062 - 1071
KRUSKAL, J. B.: "Time warps, string edits and macromolecules: the theory and practice of sequence comparison", 1983, ADDISON WESLEY, article "An overview of sequence comparison", pages: 1 - 44
KYTE ET AL., J. MOL. BIOL., vol. 157, 1982, pages 105 - 131
LARSEN ET AL., DIABETES CARE, vol. 32, 2009, pages 1663 - 8
LARSEN ET AL., NEJM, vol. 356, 2007, pages 1517 - 26
LEE, H. C. ET AL., NATURE, vol. 408, 2000, pages 483 - 8
LUST ET AL., MAYO CLIN PROC, vol. 84, no. 2, 2009, pages 114 - 122
MATTHEAKIS ET AL., PROC. NATL. ACAD. SCI. USA, vol. 91, 1994, pages 9022
MCLNDOE ET AL., PROC. NATL. ACAD. SCI. USA, vol. 96, 1999, pages 2210 - 2214
MCMAHAN ET AL., EMBO J., vol. 10, 1991, pages 2821
MEENAN ET AL., ARTHRITIS RHEUM., vol. 35, no. 1, 1992, pages 1 - 10
MYERS, MILLER, CABIOS, vol. 4, 1988, pages 11 - 17
NEEDLEMAN, S. B., WUNSCH, C. D., J. MOL. BIOL., vol. 48, 1970, pages 443 - 453
NUCLEIC ACIDS RES., 2010, pages D142 - D148
NUKI, G. ET AL., ARTHRITIS & RHEUMATISM, vol. 46, 2002, pages 2838 - 46
OCUL IMMUNOL INFLAMM., vol. 5, no. 5, September 2007 (2007-09-01), pages 389 - 93
OKA, K. ET AL., CURR. OPIN. LIPIDOL., vol. 11, 2000, pages 179 - 86
OLSON ET AL., OCUL IMMUNOL INFLAMM, vol. 17, no. 3, 2009, pages 195 - 200
OWYANG ET AL., ENDOCRINOLOGY, vol. 151, no. 6, 2010, pages 2515 - 27
PAWLIUK ET AL., SCIENCE, vol. 294, 2001, pages 2368
PEARSON, PROTEIN SCIENCE, vol. 4, 1995, pages 1145 - 1160
PINCUS ET AL., ARTHRITIS RHEUM., vol. 26, no. 11, 1983, pages 1346 - 53
QUARTIER ET AL., ANN RHEUM DIS., vol. 70, no. 5, 2011, pages 747 - 54
RAETHER: "Surface Plasmons", 1988, SPRINGER VERLAG
RICE, P. ET AL.: "EMBOSS: The European Molecular Biology Open Software Suite", TRENDS IN GENETICS, vol. 16, no. 6, 2000, pages 276 - 277, XP004200114, Retrieved from the Internet <URL:http://www.ebi.ac.uk/Tools/emboss/ align/index.html> DOI: doi:10.1016/S0168-9525(00)02024-2
ROTHWELL, N. J. ET AL., TINS, vol. 23, no. 12, 2000, pages 618 - 625
SCHAFFITZEL ET AL., J IMMUNOL METHODS., vol. 231, no. 1-2, 1999, pages 119 - 3
SCHIFF, M.H. ET AL., ARTHRITIS & RHEUMATISM, vol. 50, 2004, pages 1752 - 60
SCHREUDER ET AL., NATURE, vol. 386, 1997, pages 194 - 200
SCOPES: "Protein Purification: Principles and Practice", 2010, SPRINGER-VERLAG
SHIELDS ET AL., J. BIOL. CHEM., vol. 276, 2001, pages 6591 - 6604
SHIRATORY, Y. ET AL., LIVER, vol. 19, 1999, pages 265 - 74
SIMS, SMITH: "The IL-1 family: regulators of immunity", NATURE REVIEWS IMMUNOLOGY, 2010
SJOLANDER, URBANICZKY, ANAL. CHEM., vol. 63, 1991, pages 2338 - 2345
SMITH, SCIENCE, vol. 228, 1985, pages 1315 - 1317
SMITH-ARICA, J. R., BARTLETT, J. S., CURR. CARDIOL. REP., vol. 3, 2001, pages 43 - 49
STRAYER, D. S., EXPERT OPIN. INVESTIG. DRUGS, vol. 8, 1999, pages 2159 - 2172
SZABO ET AL., CURR. OPIN. STRUCT. BIOL., vol. 5, 1995, pages 699 - 705
THOMPSON ET AL., NUCL. ACIDS RES., vol. 28, 2000, pages 2910 - 2926
THULE, P. M., LIU, J. M., GENE THER., vol. 7, 2000, pages 1744 - 52
TINUBU ET AL., J. IMMUNOL., 1994, pages 4330 - 4338
TRITTIBACH P ET AL., GENE THER., vol. 15, no. 22, 2008, pages 1478 - 88
TSAI ET AL., MOL VIS, vol. 15, 2009, pages 1542 - 1552
URLAUB ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 4216
URLINGER, S. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 97, no. 14, 2000, pages 7963 - 7968
VIGERS ET AL., NATURE, vol. 386, 1997, pages 190 - 194
VITI ET AL., METHODS ENZYMOL., vol. 326, 2000, pages 480 - 505
VORONOV ET AL., PNAS, vol. 100, 2003, pages 2645 - 2650
WARE ET AL., MED. CARE., vol. 37, no. 5, 1999, pages MS40 - 50
WELLS ET AL., GENE, vol. 34, 1985, pages 315
WELLS ET AL., PHILOS. TRANS. R. SOC. LONDON, vol. 317, 1986, pages 415
YANG, N. S., CRIT. REV. BIOTECHNOL., vol. 12, 1992, pages 335 - 56
ZOLLER ET AL., NUCL. ACIDS RES., vol. 10, 1987, pages 6487

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9163072B2 (en) 2007-05-01 2015-10-20 Alligator Bioscience Ab Mutants of interleukin-1 receptor antagonist
US9458216B2 (en) 2010-07-29 2016-10-04 Eleven Biotherapeutics, Inc. Nucleic acid encoding chimeric IL-1 receptor type I antagonists
WO2012103240A3 (en) * 2011-01-25 2012-10-26 Eleven Biotherapeutics, Inc. Chimeric il-1 receptor type i agonists and antagonists
WO2013019652A1 (en) * 2011-07-29 2013-02-07 Eleven Biotherapeutics, Inc. Purified proteins
EP3401402A1 (en) * 2012-06-08 2018-11-14 Alkermes Pharma Ireland Limited Ligands modified by circular permutation as agonists and antagonists
AU2015268652B2 (en) * 2012-06-08 2017-11-23 Alkermes Pharma Ireland Limited Ligands modified by circular permutation as agonists and antagonists
EP2859015A4 (en) * 2012-06-08 2015-11-18 Alkermes Inc Ligands modified by circular permutation as agonists and antagonists
AU2018200597B2 (en) * 2012-06-08 2019-08-29 Alkermes Pharma Ireland Limited Ligands modified by circular permutation as agonists and antagonists
US9359415B2 (en) 2012-06-08 2016-06-07 Alkermes, Inc. Ligands modified by circular permutation as agonists and antagonists
JP2018038389A (en) * 2012-06-08 2018-03-15 アルカーメス,インコーポレイテッド Ligands modified by circular permutation as agonists and antagonists
US9428563B2 (en) 2012-06-08 2016-08-30 Alkermes, Inc. Ligands modified by circular permutation as agonists and antagonists
JP2015525217A (en) * 2012-06-08 2015-09-03 アルカーメス,インコーポレイテッド Ligands modified by circulating substitution as agonists and antagonists
JP2016521252A (en) * 2013-03-13 2016-07-21 イレブン・バイオセラピユーテイクス・インコーポレイテツド Chimeric cytokine formulations for ocular delivery
CN107693781A (en) * 2013-03-13 2018-02-16 十生物治疗股份有限公司 Chimeric cell factor formulations for ocular delivery
US20180177877A1 (en) * 2013-03-13 2018-06-28 Eleven Biotherapeutics, Inc. Chimeric cytokine formulations for ocular delivery
WO2014160371A1 (en) 2013-03-13 2014-10-02 Eleven Biotherapeutics, Inc. Chimeric cytokine formulations for ocular delivery
AU2014243839B2 (en) * 2013-03-13 2019-01-03 Buzzard Pharmaceuticals AB Chimeric cytokine formulations for ocular delivery
CN105392494A (en) * 2013-03-13 2016-03-09 十一生物治疗股份有限公司 Chimeric cytokine formulations for ocular delivery
US10799589B2 (en) 2013-03-13 2020-10-13 Buzzard Pharmaceuticals AB Chimeric cytokine formulations for ocular delivery
CN107693781B (en) * 2013-03-13 2021-11-09 巴扎德制药公司 Chimeric cytokine formulations for ocular delivery
EP3004154A4 (en) * 2013-06-05 2017-01-04 Ben Gurion University of The Negev Research and Development Authority IL-1beta-IL-1Ra CHIMERA AND METHODS OF USING THE SAME
US9895420B2 (en) 2013-06-05 2018-02-20 B.G. Negev Technologies And Applications Ltd., At Ben Gurion University IL-1beta propeptide and IL-1Ra chimera and methods of using the same
WO2015138844A1 (en) 2014-03-13 2015-09-17 Eleven Biotherapeutics, Inc. Methods for production of stable chimeric cytokine protein formulations in blow fill seal containers
WO2023169686A1 (en) 2022-03-10 2023-09-14 Buzzard Pharmaceuticals AB Inhibitor of interleukin-1 receptor type 1 for use in the treatment of cancer

Also Published As

Publication number Publication date
US9458216B2 (en) 2016-10-04
CA2806438A1 (en) 2012-02-02
EP3470421B1 (en) 2023-06-07
JP6030554B2 (en) 2016-11-24
EP4219538A2 (en) 2023-08-02
CN103221422B (en) 2017-03-29
CN103221422A (en) 2013-07-24
EP3470421C0 (en) 2023-06-07
HRP20181446T1 (en) 2018-11-02
MX360097B (en) 2018-10-22
RU2013108836A (en) 2014-09-10
HRP20230776T1 (en) 2023-10-27
HUE063115T2 (en) 2023-12-28
RU2017132694A3 (en) 2020-12-22
US20130209396A1 (en) 2013-08-15
EP2598526A1 (en) 2013-06-05
LT2598526T (en) 2018-10-10
IL224435B (en) 2018-06-28
DK2598526T3 (en) 2018-11-19
CA2806438C (en) 2021-04-13
EP3470421A1 (en) 2019-04-17
CN107337735B (en) 2021-06-22
ES2952394T3 (en) 2023-10-31
EP3470421B9 (en) 2023-10-04
AU2018250431A1 (en) 2018-11-15
AU2016213845B2 (en) 2018-07-19
KR20130136447A (en) 2013-12-12
RU2017132694A (en) 2019-02-06
SI2598526T1 (en) 2018-10-30
SG187231A1 (en) 2013-02-28
PL2598526T4 (en) 2019-02-28
IL259547A (en) 2018-07-31
BR112013003646A2 (en) 2017-07-04
KR102080704B1 (en) 2020-02-24
MX2013001107A (en) 2013-06-18
ES2701405T3 (en) 2019-02-22
BR112013003646B1 (en) 2021-09-08
HK1246327A1 (en) 2018-09-07
EP2598526B1 (en) 2018-09-05
PL3470421T3 (en) 2023-10-23
PL2598526T3 (en) 2019-02-28
US20150087818A1 (en) 2015-03-26
AU2016213845A1 (en) 2016-09-29
EP4219538A3 (en) 2023-08-09
US8853150B2 (en) 2014-10-07
JP2013535204A (en) 2013-09-12
ZA201505145B (en) 2015-09-30
CN107337735A (en) 2017-11-10
RU2754957C2 (en) 2021-09-08
AU2011283669A1 (en) 2013-02-07
HUE041590T2 (en) 2019-05-28
NZ605928A (en) 2015-02-27
IL259547B (en) 2020-04-30

Similar Documents

Publication Publication Date Title
AU2016213845B2 (en) Chimeric il-1 receptor type i agonists and antagonists
US20140341843A1 (en) Purified proteins
WO2012103240A2 (en) Receptor binding agents
US20190300601A1 (en) Correctly folded etanercept in high purity and excellent yield
EP2456787A2 (en) Cytokine compositions and methods of use thereof
JP7306655B2 (en) IL-37 variant
US20040204351A1 (en) Soluble proteins that inhibit cytokine signal transduction pathways
TWI586803B (en) Chimeric il-1 receptor type i antagonists
US20140105855A1 (en) Structural based design of il-17 dominant negative mutants

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11749612

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2806438

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2013522011

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 224435

Country of ref document: IL

Ref document number: MX/A/2013/001107

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2011283669

Country of ref document: AU

Date of ref document: 20110729

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2011749612

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2013108836

Country of ref document: RU

Kind code of ref document: A

Ref document number: 20137005337

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13812582

Country of ref document: US

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013003646

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013003646

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20130128